1
|
VanNatta JM, Niu H, Carlson GJ, Pinney KG. Application of Chlorosulfonyl Isocyanate (CSI) in the Synthesis of Fused Tetracyclic Ketone Ring Systems. J Org Chem 2024; 89:15636-15651. [PMID: 39388523 DOI: 10.1021/acs.joc.4c01714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Chlorosulfonyl isocyanate (CSI) is a complex reagent capable of facilitating numerous synthetic transformations, including lactam/lactone formation, sulfonylation, Friedel-Crafts-type acylations, and cycloadditions. Annulation reactions to form nitrogen-, oxygen-, and sulfur-bearing heterocycles have been observed with CSI; however, the application of CSI toward the generation of fused cyclic ketone ring systems has not been previously reported. A serendipitous discovery of the pertinence of CSI occurred during a structure-activity relationship campaign around our established lead benzosuberene-based molecule that functions as a potent inhibitor of tubulin polymerization. The benzylic olefin within this molecule represents a promising moiety for further functionalization. CSI was initially investigated as a reagent to effect transformation of this olefin to its corresponding β-lactam functionality, but instead resulted in an unexpected tetracyclic fused ring system in high yield (88%). This finding led to an exploration of the reactivity of CSI with various arenes. Benzosuberene analogues with varying functionalizations were synthesized and treated with CSI, with all examples resulting in a fused ring system except those bearing electron-withdrawing groups. Notably, simplified arene structures with fewer substituents were also observed to undergo cyclization under these conditions. This strategy represents a promising approach for the synthesis of appropriately functionalized tetracyclic ring systems.
Collapse
Affiliation(s)
- Jennifer M VanNatta
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Haichan Niu
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Graham J Carlson
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, Texas 76798-7348, United States
| |
Collapse
|
2
|
Ozcan BB, Wanniarachchi H, Mason RP, Dogan BE. Current status of optoacoustic breast imaging and future trends in clinical application: is it ready for prime time? Eur Radiol 2024; 34:6092-6107. [PMID: 38308678 PMCID: PMC11297194 DOI: 10.1007/s00330-024-10600-2] [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: 09/08/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 02/05/2024]
Abstract
Optoacoustic imaging (OAI) is an emerging field with increasing applications in patients and exploratory clinical trials for breast cancer. Optoacoustic imaging (or photoacoustic imaging) employs non-ionizing, laser light to create thermoelastic expansion in tissues and detect the resulting ultrasonic emission. By combining high optical contrast capabilities with the high spatial resolution and anatomic detail of grayscale ultrasound, OAI offers unique opportunities for visualizing biological function of tissues in vivo. Over the past decade, human breast applications of OAI, including benign/malignant mass differentiation, distinguishing cancer molecular subtype, and predicting metastatic potential, have significantly increased. We discuss the current state of optoacoustic breast imaging, as well as future opportunities and clinical application trends. CLINICAL RELEVANCE STATEMENT: Optoacoustic imaging is a novel breast imaging technique that enables the assessment of breast cancer lesions and tumor biology without the risk of ionizing radiation exposure, intravenous contrast, or radionuclide injection. KEY POINTS: • Optoacoustic imaging (OAI) is a safe, non-invasive imaging technique with thriving research and high potential clinical impact. • OAI has been considered a complementary tool to current standard breast imaging techniques. • OAI combines parametric maps of molecules that absorb light and scatter acoustic waves (like hemoglobin, melanin, lipids, and water) with anatomical images, facilitating scalable and real-time molecular evaluation of tissues.
Collapse
Affiliation(s)
- B Bersu Ozcan
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard MC 8896, Dallas, TX, 75390-8896, USA.
| | - Hashini Wanniarachchi
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard MC 8896, Dallas, TX, 75390-8896, USA
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard MC 8896, Dallas, TX, 75390-8896, USA
| | - Basak E Dogan
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard MC 8896, Dallas, TX, 75390-8896, USA
| |
Collapse
|
3
|
Hoyt K. Super-Resolution Ultrasound Imaging for Monitoring the Therapeutic Efficacy of a Vascular Disrupting Agent in an Animal Model of Breast Cancer. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1099-1107. [PMID: 38411352 DOI: 10.1002/jum.16438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVE Evaluate the use of super-resolution ultrasound (SRUS) imaging for the early detection of tumor response to treatment using a vascular-disrupting agent (VDA). METHODS A population of 28 female nude athymic mice (Charles River Laboratories) were implanted with human breast cancer cells (MDA-MB-231, ATCC) in the mammary fat pad and allowed to grow. Ultrasound imaging was performed using a Vevo 3100 scanner (FUJIFILM VisualSonics Inc) equipped with the MX250 linear array transducer immediately before and after receiving bolus injections of a microbubble (MB) contrast agent (Definity, Lantheus Medical Imaging) via the tail vein. Following baseline ultrasound imaging, VDA drug (combretastatin A4 phosphate, CA4P, Sigma Aldrich) or control saline was injected via the placed catheter. After 4 or 24 hours, repeat ultrasound imaging along the same tumor cross-section occurred. Direct intratumoral pressure measurements were obtained using a calibrated sensor. All raw ultrasound data were saved for offline processing and SRUS image reconstruction using custom MATLAB software (MathWorks Inc). From a region encompassing the tumor space and the entire postprocessed ultrasound image sequence, time MB count (TMC) curves were generated in addition to traditional SRUS maps reflecting MB enumeration at each pixel location. Peak enhancement (PE) and wash-in rate (WIR) were extracted from these TMC curves. At termination, intratumoral microvessel density (MVD) was quantified using tomato lectin labeling of patent blood vessels. RESULTS SRUS images exhibited a clear difference between control and treated tumors. While there was no difference in any group parameters at baseline (0 hour, P > .09), both SRUS-derived PE and WIR measurements in tumors treated with VDA exhibited significant decreases by 4 (P = .03 and P = .05, respectively) and 24 hours (P = .02 and P = .01, respectively), but not in control group tumors (P > .22). Similarly, SRUS derived microvascular maps were not different at baseline (P = .81), but measures of vessel density were lower in treated tumors at both 4 and 24 hours (P < .04). An inverse relationship between intratumoral pressure and both PE and WIR parameters were found in control tumors (R2 > .09, P < .03). CONCLUSION SRUS imaging is a new modality for assessing tumor response to treatment using a VDA.
Collapse
Affiliation(s)
- Kenneth Hoyt
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
4
|
Ren W, Deng Y, Ward JD, Vairin R, Bai R, Wanniarachchi HI, Hamal KB, Tankoano PE, Tamminga CS, Bueno LMA, Hamel E, Mason RP, Trawick ML, Pinney KG. Synthesis and biological evaluation of structurally diverse 6-aryl-3-aroyl-indole analogues as inhibitors of tubulin polymerization. Eur J Med Chem 2024; 263:115794. [PMID: 37984295 PMCID: PMC11019941 DOI: 10.1016/j.ejmech.2023.115794] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 11/22/2023]
Abstract
The synthesis and evaluation of small-molecule inhibitors of tubulin polymerization remains a promising approach for the development of new therapeutic agents for cancer treatment. The natural products colchicine and combretastatin A-4 (CA4) inspired significant drug discovery campaigns targeting the colchicine site located on the beta-subunit of the tubulin heterodimer, but so far these efforts have not yielded an approved drug for cancer treatment in human patients. Interest in the colchicine site was enhanced by the discovery that a subset of colchicine site agents demonstrated dual functionality as both potent antiproliferative agents and effective vascular disrupting agents (VDAs). Our previous studies led to the discovery and development of a 2-aryl-3-aroyl-indole analogue (OXi8006) that inhibited tubulin polymerization and demonstrated low nM IC50 values against a variety of human cancer cell lines. A water-soluble phosphate prodrug salt (OXi8007), synthesized from OXi8006, displayed promising vascular disrupting activity in mouse models of cancer. To further extend structure-activity relationship correlations, a series of 6-aryl-3-aroyl-indole analogues was synthesized and evaluated for their inhibition of tubulin polymerization and cytotoxicity against human cancer cell lines. Several structurally diverse molecules in this small library were strong inhibitors of tubulin polymerization and of MCF-7 and MDA-MB-231 human breast cancer cells. One of the most promising analogues (KGP591) caused significant G2/M arrest of MDA-MB-231 cells, disrupted microtubule structure and cell morphology in MDA-MB-231 cells, and demonstrated significant inhibition of MDA-MB-231 cell migration in a wound healing (scratch) assay. A phosphate prodrug salt, KGP618, synthesized from its parent phenolic precursor, KGP591, demonstrated significant reduction in bioluminescence signal when evaluated in vivo against an orthotopic model of kidney cancer (RENCA-luc) in BALB/c mice, indicative of VDA efficacy. The most active compounds from this series offer promise as anticancer therapeutic agents.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Hashini I Wanniarachchi
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Khagendra B Hamal
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Caleb S Tamminga
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Lorena M A Bueno
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| |
Collapse
|
5
|
Ren W, Vairin R, Ward JD, Francis R, VanNatta J, Bai R, Tankoano PE, Deng Y, Hamel E, Trawick ML, Pinney KG. Structure Guided Design, Synthesis, and Biological Evaluation of Oxetane-Containing Indole Analogues. Bioorg Med Chem 2023; 92:117400. [PMID: 37556912 PMCID: PMC10848874 DOI: 10.1016/j.bmc.2023.117400] [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: 05/20/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023]
Abstract
The oxetane functional group offers a variety of potential advantages when incorporated within appropriate therapeutic agents as a ketone surrogate. OXi8006, a 2-aryl-3-aroyl-indole analogue, functions as a small-molecule inhibitor of tubulin polymerization that has a dual mechanism of action as both an antiproliferative agent and a tumor-selective vascular disrupting agent. Replacement of the bridging ketone moiety in OXi8006 with an oxetane functional group has expanded structure activity relationship (SAR) knowledge and provided insights regarding oxetane incorporation within this class of molecules. A new synthetic method using an oxetane-containing tertiary alcohol subjected to Lewis acid catalyzed conditions led to successful Friedel-Crafts alkylation and yielded fourteen new oxetane-containing indole-based molecules. This synthetic approach represents the first method to successfully install an oxetane ring at the 3-position of a 2-aryl-indole system. Several analogues showed potent cytotoxicity (micromolar GI50 values) against human breast cancer cell lines (MCF-7 and MDA-MB-231) and a pancreatic cancer cell line (PANC-1), although they proved to be ineffective as inhibitors of tubulin polymerization. Molecular docking studies comparing colchicine with the OXi8006-oxetane analogue 5m provided a rationale for the differential interaction of these molecules with the colchicine site on the tubulin heterodimer.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ricardo Francis
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jenny VanNatta
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States.
| |
Collapse
|
6
|
Kagalwala HN, Bueno L, Wanniarachchi H, Unruh DK, Hamal KB, Pavlich CI, Carlson GJ, Pinney KG, Mason RP, Lippert AR. Oxygen-Sensing Chemiluminescent Iridium(III) 1,2-Dioxetanes: Unusual Coordination and Activity. ANALYSIS & SENSING 2023; 3:e202200085. [PMID: 37006671 PMCID: PMC10061878 DOI: 10.1002/anse.202200085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Indexed: 11/23/2022]
Abstract
Next generation chemiluminescent iridium 1,2-dioxetane complexes have been developed which consist of the Schaap's 1,2-dioxetane scaffold directly attached to the metal center. This was achieved by synthetically modifying the scaffold precursor with a phenylpyridine moiety, which can act as a ligand. Reaction of this scaffold ligand with the iridium dimer [Ir(BTP)2(μ-Cl)]2 (BTP = 2-(benzo[b]thiophen-2-yl)pyridine) yielded isomers which depict ligation through either the cyclometalating carbon or, interestingly, the sulfur atom of one BTP ligand. Their corresponding 1,2-dioxetanes display chemiluminescent responses in buffered solutions, exhibiting a single, red-shifted peak at 600 nm. This triplet emission was effectively quenched by oxygen, yielding in vitro Stern-Volmer constants of 0.1 and 0.009 mbar-1 for the carbon-bound and sulfur compound, respectively. Lastly, the sulfur-bound dioxetane was further utilized for oxygen sensing in muscle tissue of living mice and xenograft models of tumor hypoxia, depicting the ability of the probe chemiluminescence to penetrate biological tissue (total flux ~ 106 p/s).
Collapse
Affiliation(s)
- Husain N Kagalwala
- Department of Chemistry, Southern Methodist University, Dallas, TX 75275-0314 (USA)
| | - Lorena Bueno
- Prognostic Imaging Research Laboratory, Pre-clinical Imaging Section, Department of Radiology, UT Southwestern Medical Center, Dallas, TX 75390-9058 (USA)
| | - Hashini Wanniarachchi
- Prognostic Imaging Research Laboratory, Pre-clinical Imaging Section, Department of Radiology, UT Southwestern Medical Center, Dallas, TX 75390-9058 (USA)
| | - Daniel K Unruh
- X-ray Diffraction Facility, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061 (USA)
| | - Khagendra B Hamal
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798-7348 (USA)
| | - Cyprian I Pavlich
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798-7348 (USA)
| | - Graham J Carlson
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798-7348 (USA)
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798-7348 (USA)
| | - Ralph P Mason
- Prognostic Imaging Research Laboratory, Pre-clinical Imaging Section, Department of Radiology, UT Southwestern Medical Center, Dallas, TX 75390-9058 (USA)
| | - Alexander R Lippert
- Department of Chemistry, Southern Methodist University, Dallas, TX 75275-0314 (USA)
| |
Collapse
|
7
|
Sun R, Xiang J, Zhou Q, Piao Y, Tang J, Shao S, Zhou Z, Bae YH, Shen Y. The tumor EPR effect for cancer drug delivery: Current status, limitations, and alternatives. Adv Drug Deliv Rev 2022; 191:114614. [PMID: 36347432 DOI: 10.1016/j.addr.2022.114614] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2022]
Abstract
Over the past three decades, the enhanced permeability and retention (EPR) effect has been considered the basis of tumor-targeted drug delivery. Various cancer nanomedicines, including macromolecular drugs, have been designed to utilize this mechanism for preferential extravasation and accumulation in solid tumors. However, such nanomedicines have not yet achieved convincing therapeutic benefits in clinics. Increasing evidence suggests that the EPR effect is over-represented in human tumors, especially in metastatic tumors. This review covers the evolution of the concept, the heterogeneity and limitation of the EPR effect in clinical realities, and prospects for alternative strategies independent of the EPR effect.
Collapse
Affiliation(s)
- Rui Sun
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - You Han Bae
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
8
|
Mampaey G, Hellemans A, de Rooster H, Schipper T, Abma E, Broeckx BJG, Daminet S, Smets P. Assessment of Cardiotoxicity after a Single Dose of Combretastatin A4-Phosphate in Dogs Using Two-Dimensional Speckle-Tracking Echocardiography. Animals (Basel) 2022; 12:ani12213005. [PMID: 36359129 PMCID: PMC9658292 DOI: 10.3390/ani12213005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/21/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Simple Summary Combretastatin A4-phosphate is a chemotherapeutic drug which has been evaluated for treatment of solid canine tumors. Previous studies reported cardiotoxic effects based on changes in cardiac troponin I measurements, blood pressure, and electrocardiography. We evaluated the cardiotoxic effect by two-dimensional speckle tracking echocardiography. This advanced imaging technique analyzes global and regional myocardial function and is used as the gold-standard for the assessment of cardiac function in human patients receiving chemotherapy. We found that certain strain measurements were significantly decreased 24 h after the administration of combretastatin A4-phosphate and that these changes were correlated with an increase in cardiac troponin I. Our results suggest that two-dimensional speckle tracking may be useful for the early detection of cardiac dysfunction in canine cancer patients as well as promising during follow-up. Abstract Combretastatin A4-phosphate (CA4P) is a vascular disrupting agent that was recently described for the treatment of solid canine tumors. Conventional echocardiography and pulsed wave tissue Doppler imaging did not reveal cardiotoxicity in dogs, however, the gold standard for assessing myocardial damage in humans receiving cardiotoxic chemotherapeutics is two-dimensional speckle-tracking echocardiography. The current study evaluated the cardiotoxic effect of a single dose of CA4P in dogs using peak systolic strain measurements and the variability of these measurements. Echocardiographic examinations of seven healthy beagles and five canine cancer patients that received CA4P were retrospectively reviewed. Peak systolic regional longitudinal strain (LSt), peak systolic regional circumferential strain (CSt), and peak systolic regional radial strain (RSt) were measured before and 24 h after administration of CA4P. Peak systolic strain measurements were compared to serum cardiac troponin I (cTnI). To quantify intra- and inter-observer measurement variability, seven echocardiographic examinations were selected and each strain parameter was measured by three observers on three consecutive days. After CA4P administration, the median LSt and CSt values decreased by 21.8% (p = 0.0005) and 12.3% (p = 0.002), respectively, whereas the median RSt values were not significantly different (p = 0.70). The decrease in LSt was correlated with increased serum cTnI values (Spearman rho = −0.64, p = 0.02). The intra-observer coefficients of variation (CV) were 9%, 4%, and 13% for LSt, CSt, and RSt, respectively, while the corresponding interobserver CVs were 11%, 12%, and 20%. Our results suggest that regional peak systolic strain measurements may be useful for the early detection of cardiotoxicity that is caused by vascular disrupting agents and that LSt may be promising for the follow-up of canine cancer patients.
Collapse
Affiliation(s)
- Gitte Mampaey
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
- Correspondence:
| | - Arnaut Hellemans
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Tom Schipper
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Bart J. G. Broeckx
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Sylvie Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Pascale Smets
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| |
Collapse
|
9
|
Liu L, Schuetze R, Gerberich JL, Lopez R, Odutola SO, Tanpure RP, Charlton-Sevcik AK, Tidmore JK, Taylor EAS, Kapur P, Hammers H, Trawick ML, Pinney KG, Mason RP. Demonstrating Tumor Vascular Disrupting Activity of the Small-Molecule Dihydronaphthalene Tubulin-Binding Agent OXi6196 as a Potential Therapeutic for Cancer Treatment. Cancers (Basel) 2022; 14:4208. [PMID: 36077745 PMCID: PMC9454770 DOI: 10.3390/cancers14174208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The vascular disrupting activity of a promising tubulin-binding agent (OXi6196) was demonstrated in mice in MDA-MB-231 human breast tumor xenografts growing orthotopically in mammary fat pad and syngeneic RENCA kidney tumors growing orthotopically in the kidney. To enhance water solubility, OXi6196, was derivatized as its corresponding phosphate prodrug salt OXi6197, facilitating effective delivery. OXi6197 is stable in water, but rapidly releases OXi6196 in the presence of alkaline phosphatase. At low nanomolar concentrations OXi6196 caused G2/M cell cycle arrest and apoptosis in MDA-MB-231 breast cancer cells and monolayers of rapidly growing HUVECs underwent concentration-dependent changes in their morphology. Loss of the microtubule structure and increased bundling of filamentous actin into stress fibers followed by cell collapse, rounding and blebbing was observed. OXi6196 (100 nM) disrupted capillary-like endothelial networks pre-established with HUVECs on Matrigel®. When prodrug OXi6197 was administered to mice bearing orthotopic MDA-MB-231-luc tumors, dynamic bioluminescence imaging (BLI) revealed dose-dependent vascular shutdown with >80% signal loss within 2 h at doses ≥30 mg/kg and >90% shutdown after 6 h for doses ≥35 mg/kg, which remained depressed by at least 70% after 24 h. Twice weekly treatment with prodrug OXi6197 (20 mg/kg) caused a significant tumor growth delay, but no overall survival benefit. Similar efficacy was observed for the first time in orthotopic RENCA-luc tumors, which showed massive hemorrhage and necrosis after 24 h. Twice weekly dosing with prodrug OXi6197 (35 mg/kg) caused tumor growth delay in most orthotopic RENCA tumors. Immunohistochemistry revealed extensive necrosis, though with surviving peripheral tissues. These results demonstrate effective vascular disruption at doses comparable to the most effective vascular-disrupting agents (VDAs) suggesting opportunities for further development.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Regan Schuetze
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeni L. Gerberich
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ramona Lopez
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel O. Odutola
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Rajendra P. Tanpure
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | | | - Justin K. Tidmore
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Emily A.-S. Taylor
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Payal Kapur
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hans Hammers
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
10
|
Wadsworth BJ, Lee CM, Bennewith KL. Transiently hypoxic tumour cell turnover and radiation sensitivity in human tumour xenografts. Br J Cancer 2022; 126:1616-1626. [PMID: 35031765 PMCID: PMC9130130 DOI: 10.1038/s41416-021-01691-5] [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: 09/14/2020] [Revised: 11/24/2021] [Accepted: 12/23/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Solid tumour perfusion can be unstable, creating transiently hypoxic cells that can contribute to radiation resistance. We investigated the in vivo lifetime of transiently hypoxic tumour cells and chronically hypoxic tumour cells during tumour growth and following irradiation. METHODS Hypoxic cells in SiHa and WiDr human tumour xenografts were labelled using pimonidazole and EF5, and turnover was quantified as the loss of labelled cells over time. The perfusion-modifying drug pentoxifylline was used to reoxygenate transiently hypoxic cells prior to hypoxia marker administration or irradiation. RESULTS Chronically hypoxic cells constantly turnover in SiHa and WiDr tumours, with half-lives ranging from 42-82 h and significant numbers surviving >96 h. Transiently hypoxic cells constitute 26% of the total hypoxic cells in WiDr tumours. These transiently hypoxic cells survive at least 24 h, but then rapidly turnover with a half-life of 34 h and are undetectable 72 h after labelling. Transiently hypoxic cells are radiation-resistant, although vascular dysfunction induced by 10 Gy of ionising radiation preferentially kills transiently hypoxic cells. CONCLUSIONS Transiently hypoxic tumour cells survive up to 72 h in WiDr tumours and are radiation-resistant, although transiently hypoxic cells are sensitive to vascular dysfunction induced by high doses of ionising radiation.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Che-Min Lee
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
11
|
Guo Y, Wang H, Gerberich JL, Odutola SO, Charlton-Sevcik AK, Li M, Tanpure RP, Tidmore JK, Trawick ML, Pinney KG, Mason RP, Liu L. Imaging-Guided Evaluation of the Novel Small-Molecule Benzosuberene Tubulin-Binding Agent KGP265 as a Potential Therapeutic Agent for Cancer Treatment. Cancers (Basel) 2021; 13:cancers13194769. [PMID: 34638255 PMCID: PMC8507561 DOI: 10.3390/cancers13194769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Vascular-disrupting agents promise significant therapeutic efficacy against solid tumors by selectively damaging tumor-associated vasculature. Dynamic BLI and oxygen-enhanced multispectral optoacoustic tomography (OE-MSOT) were used to compare vascular shutdown following administration of KGP265. BLI signal and vascular oxygenation response (ΔsO2) to a gas breathing challenge were both significantly reduced within 2 h indicating vascular disruption, which continued over 24 h. Twice-weekly doses of KGP265 caused a significant growth delay in MDA-MB-231 human breast tumor xenografts and 4T1 syngeneic breast tumors growing orthotopically in mice. Abstract The selective disruption of tumor-associated vasculature represents an attractive therapeutic approach. We have undertaken the first in vivo evaluation of KGP265, a water-soluble prodrug of a benzosuberene-based tubulin-binding agent, and found promising vascular-disrupting activity in three distinct tumor types. Dose escalation in orthotopic MDA-MB-231-luc breast tumor xenografts in mice indicated that higher doses produced more effective vascular shutdown, as revealed by dynamic bioluminescence imaging (BLI). In syngeneic orthotopic 4T1-luc breast and RENCA-luc kidney tumors, dynamic BLI and oxygen enhanced multispectral optoacoustic tomography (OE-MSOT) were used to compare vascular shutdown following the administration of KGP265 (7.5 mg/kg). The BLI signal and vascular oxygenation response (ΔsO2) to a gas breathing challenge were both significantly reduced within 2 h, indicating vascular disruption, which continued over 24 h. A correlative histology confirmed increased necrosis and hemorrhage. Twice-weekly doses of KGP265 caused significant growth delay in both MDA-MB-231 and 4T1 breast tumors, with no obvious systemic toxicity. A combination with carboplatin produced significantly greater tumor growth delay than carboplatin alone, though significant carboplatin-associated toxicity was observed (whole-body weight loss). KGP265 was found to be effective at low concentrations, generating long-term vascular shutdown and tumor growth delay, thus providing strong rationale for further development, particularly in combination therapies.
Collapse
Affiliation(s)
- Yihang Guo
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha 410013, China
| | - Honghong Wang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jeni L. Gerberich
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
| | - Samuel O. Odutola
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Amanda K. Charlton-Sevcik
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Maoping Li
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rajendra P. Tanpure
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Justin K. Tidmore
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (R.P.M.); (L.L.)
| | - Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (R.P.M.); (L.L.)
| |
Collapse
|
12
|
The Proper Administration Sequence of Radiotherapy and Anti-Vascular Agent-DMXAA Is Essential to Inhibit the Growth of Melanoma Tumors. Cancers (Basel) 2021; 13:cancers13163924. [PMID: 34439079 PMCID: PMC8394873 DOI: 10.3390/cancers13163924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023] Open
Abstract
Vascular disrupting agents (VDAs), such as DMXAA, effectively destroy tumor blood vessels and cause the formation of large areas of necrosis in the central parts of the tumors. However, the use of VDAs is associated with hypoxia activation and residues of rim cells on the edge of the tumor that are responsible for tumor regrowth. The aim of the study was to combine DMXAA with radiotherapy (brachytherapy) and find the appropriate administration sequence to obtain the maximum synergistic therapeutic effect. We show that the combination in which tumors were irradiated prior to VDAs administration is more effective in murine melanoma growth inhibition than in either of the agents individually or in reverse combination. For the first time, the significance of immune cells' activation in such a combination is demonstrated. The inhibition of tumor growth is linked to the reduction of tumor blood vessels, the increased infiltration of CD8+ cytotoxic T lymphocytes and NK cells and the polarization of macrophages to the cytotoxic M1 phenotype. The reverse combination of therapeutic agents showed no therapeutic effect and even abolished the effect of DMXAA. The combination of brachytherapy and vascular disrupting agent effectively inhibits the growth of melanoma tumors but requires careful planning of the sequence of administration of the agents.
Collapse
|