1
|
Zhao YC, Yan LQ, Xu Y. Recent advances of selenized tubulin inhibitors in cancer therapy. Bioorg Med Chem Lett 2025; 116:130037. [PMID: 39581555 DOI: 10.1016/j.bmcl.2024.130037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Cancer treatment always a huge challenge amidst the resistance and relapse caused by the various treatments. Inhibitors targeting mitosis have been considered as promising therapeutic drugs in clinic, of which tubulins play an important role. Selenium (Se) as an essential microelement in humans and animals, playing a crucial role in the formation of anti-oxidase (glutathione peroxidase) and selenoprotein, also attracted broad attention in cancer therapy. Because the introduction of Se atom could change the length and angle of chemical bond and alter their functional properties, regulating selenized chemotherapeutics has become one of the hot spots. However, little attention has been paid to studying the combination of Se and tubulin inhibitors. Herein, we review the latest research results of selenized tubulin inhibitors in cancer therapy, including its mechanisms, categories and biological activities, providing a theoretical basis for different selenized microtubules inhibitors therapies.
Collapse
Affiliation(s)
- Yong-Chang Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Liang-Qing Yan
- Department of Radiology, The People's Hospital of Yuhuan, Taizhou 317600, China
| | - Yuan Xu
- Department of Pharmacy, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China.
| |
Collapse
|
2
|
Omar MH, Emam SH, Mikhail DS, Elmeligie S. Combretastatin A-4 based compounds as potential anticancer agents: A review. Bioorg Chem 2024; 153:107930. [PMID: 39504638 DOI: 10.1016/j.bioorg.2024.107930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
The current review discusses the importance of combretastatin A-4 (CA-4) as a lead compound of microtubule targeting agents. CA-4 holds a unique place among naturally occurring compounds having cytotoxic activity. In this review an overall picture of design strategies, structure-activity relationship, synthesis, cytotoxic activity, and binding interactions of promising CA-4 analogues, are discussed and arranged chronologically from 2016 to early 2023. Also, this review emphasizes their biological activity as anticancer agents, within an overview of clinical application limitation and suggested strategies to overcome. Dual targeting tubulin inhibitors showed highpotentialto surpass medication resistance and provide synergistic efficacy. Linking platinum (IV), amino acids, and HDAC targeting moieties to active tubulin inhibitorsproduced potent active compounds. Analogues of CA-4 bridged with azetidin-2-one, pyrazole, sulfide, or carrying selenium atom exhibited cytotoxic action against a variety of malignant cell lines through different pathways.
Collapse
Affiliation(s)
- Mai H Omar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Soha H Emam
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Demiana S Mikhail
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Salwa Elmeligie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
3
|
Vairin R, Tamminga C, Shi Z, Borchardt C, Jambulapati J, Bai R, Wanniarachchi H, Bueno L, Hamel E, Mason RP, Trawick ML, Pinney KG. Design, Synthesis and Biological Evaluation of 2-Phenyl Indole Analogues of OXi8006 as Colchicine Site Inhibitors of Tubulin Polymerization and Vascular Disrupting Agents. Bioorg Med Chem 2024; 118:117981. [PMID: 39667146 DOI: 10.1016/j.bmc.2024.117981] [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: 09/20/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 12/14/2024]
Abstract
Inhibitors of tubulin polymerization represent a promising therapeutic approach for the treatment of solid tumors. Molecules that bind to the colchicine site are of interest as they can function with a dual mechanism of action as both potent antiproliferative agents and tumor-selective vascular disrupting agents (VDAs). One such example is a 2-aryl-3-aroyl-indole molecule (OXi8006) from our laboratory that demonstrates potent inhibition of tubulin polymerization and strong antiproliferative activity (cytotoxicity) against a variety of human cancer cell lines. A water-soluble prodrug OXi8007, synthesized from OXi8006, demonstrates in vivo disruption of tumor-associated microvessels in several tumor types (mouse models). The molecular framework of OXi8006 inspired a series of fourteen new 2-aryl-3-aroyl-indole analogues that incorporated various functional group modifications on both the indole core and the aroyl ring. Electron withdrawing and donating groups at the mono-substituted 3' position and the di-substituted 3',5' positions were all accommodated while maintaining inhibition of tubulin polymerization (IC50 < 5 μM), with several analogues demonstrating activity comparable to OXi8006 and the benchmark natural product combretastatin A-4 (CA4). Preliminary structure-activity relationship (SAR) studies were further enhanced by molecular docking to predict possible colchicine site interactions. Two analogues (KGP366 and KGP369) previously synthesized in our laboratory were re-synthesized using a somewhat modified route to increase synthetic efficiency and were subsequently converted to their corresponding water-soluble phosphate prodrug salts to evaluate their efficacy as VDAs. Administration of the prodrug salt (KGP415) of KGP369 caused significant reduction in bioluminescence signal from an orthotopic kidney tumor (RENCA-luc) in BALB/c mice, indicative of VDA activity. Collectively, these new functionalized indole-based analogues have extended SAR knowledge related to the colchicine binding site, and the most biologically active analogues hold promise for continued development as pre-clinical candidates for cancer therapy.
Collapse
Affiliation(s)
- Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Caleb Tamminga
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Zhe Shi
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Christian Borchardt
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jayaram Jambulapati
- 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 Wanniarachchi
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9058, United States
| | - Lorena 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
|
4
|
Ciepła J, Smolarczyk R. Tumor hypoxia unveiled: insights into microenvironment, detection tools and emerging therapies. Clin Exp Med 2024; 24:235. [PMID: 39361163 PMCID: PMC11449960 DOI: 10.1007/s10238-024-01501-1] [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: 06/17/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Hypoxia is one of the defining characteristics of the tumor microenvironment (TME) in solid cancers. It has a major impact on the growth and spread of malignant cells as well as their resistance to common treatments like radiation and chemotherapy. Here, we explore the complex functions of hypoxia in the TME and investigate its effects on angiogenesis, immunological evasion, and cancer cell metabolism. For prognostic and therapeutic reasons, hypoxia identification is critical, and recent developments in imaging and molecular methods have enhanced our capacity to precisely locate underoxygenated areas inside tumors. Furthermore, targeted therapies that take advantage of hypoxia provide a potential new direction in the treatment of cancer. Therapeutic approaches that specifically target hypoxic conditions in tumors without causing adverse effects are being led by hypoxia-targeted nanocarriers and hypoxia-activated prodrugs (HAPs). This review provides an extensive overview of this dynamic and clinically significant area of oncology research by synthesizing current knowledge about the mechanisms of hypoxia in cancer, highlighting state-of-the-art detection methodologies, and assessing the potential and efficacy of hypoxia-targeted therapies.
Collapse
Affiliation(s)
- Joanna Ciepła
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| |
Collapse
|
5
|
Guelfi S, Hodivala-Dilke K, Bergers G. Targeting the tumour vasculature: from vessel destruction to promotion. Nat Rev Cancer 2024; 24:655-675. [PMID: 39210063 DOI: 10.1038/s41568-024-00736-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
As angiogenesis was recognized as a core hallmark of cancer growth and survival, several strategies have been implemented to target the tumour vasculature. Yet to date, attempts have rarely been so diverse, ranging from vessel growth inhibition and destruction to vessel normalization, reprogramming and vessel growth promotion. Some of these strategies, combined with standard of care, have translated into improved cancer therapies, but their successes are constrained to certain cancer types. This Review provides an overview of these vascular targeting approaches and puts them into context based on our subsequent improved understanding of the tumour vasculature as an integral part of the tumour microenvironment with which it is functionally interlinked. This new knowledge has already led to dual targeting of the vascular and immune cell compartments and sets the scene for future investigations of possible alternative approaches that consider the vascular link with other tumour microenvironment components for improved cancer therapy.
Collapse
Affiliation(s)
- Sophie Guelfi
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK.
| | - Gabriele Bergers
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Zhang Q, Xia Y, Wang L, Wang Y, Bao Y, Zhao GS. Targeted anti-angiogenesis therapy for advanced osteosarcoma. Front Oncol 2024; 14:1413213. [PMID: 39252946 PMCID: PMC11381227 DOI: 10.3389/fonc.2024.1413213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
To date, despite extensive research, the prognosis of advanced osteosarcoma has not improved significantly. Thus, patients experience a reduced survival rate, suggesting that a reevaluation of current treatment strategies is required. Recently, in addition to routine surgery, chemotherapy and radiotherapy, researchers have explored more effective and safer treatments, including targeted therapy, immunotherapy, anti-angiogenesis therapy, metabolic targets therapy, and nanomedicine therapy. The tumorigenesis and development of osteosarcoma is closely related to angiogenesis. Thus, anti-angiogenesis therapy is crucial to treat osteosarcoma; however, recent clinical trials found that it has insufficient efficacy. To solve this problem, the causes of treatment failure and improve treatment strategies should be investigated. This review focuses on summarizing the pathophysiological mechanisms of angiogenesis in osteosarcoma and recent advances in anti-angiogenesis treatment of osteosarcoma. We also discuss some clinical studies, with the aim of providing new ideas to improve treatment strategies for osteosarcoma and the prognosis of patients.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Pain and Rehabilitation, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuxuan Xia
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - LiYuan Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Sheng Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Sidorenko V, Scodeller P, Uustare A, Ogibalov I, Tasa A, Tshubrik O, Salumäe L, Sugahara KN, Simón-Gracia L, Teesalu T. Targeting vascular disrupting agent-treated tumor microenvironment with tissue-penetrating nanotherapy. Sci Rep 2024; 14:17513. [PMID: 39080306 PMCID: PMC11289491 DOI: 10.1038/s41598-024-64610-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/11/2024] [Indexed: 08/02/2024] Open
Abstract
Cancer treatment with vascular disrupting agents (VDAs) causes rapid and extensive necrosis in solid tumors. However, these agents fall short in eliminating all malignant cells, ultimately leading to tumor regrowth. Here, we investigated whether the molecular changes in the tumor microenvironment induced by VDA treatment sensitize the tumors for secondary nanotherapy enhanced by clinical-stage tumor penetrating peptide iRGD. Treatment of peritoneal carcinomatosis (PC) and breast cancer mice with VDA combretastatin A-4 phosphate (CA4P) resulted in upregulation of the iRGD receptors αv-integrins and NRP-1, particularly in the peripheral tumor tissue. In PC mice treated with CA4P, coadministration of iRGD resulted in an approximately threefold increase in tumor accumulation and a more homogenous distribution of intraperitoneally administered nanoparticles. Notably, treatment with a combination of CA4P, iRGD, and polymersomes loaded with a novel anthracycline Utorubicin (UTO-PS) resulted in a significant decrease in the overall tumor burden in PC-bearing mice, while avoiding overt toxicities. Our results indicate that VDA-treated tumors can be targeted therapeutically using iRGD-potentiated nanotherapy and warrant further studies on the sequential targeting of VDA-induced molecular signatures.
Collapse
Affiliation(s)
- Valeria Sidorenko
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, 50411, Tartu, Estonia
| | - Pablo Scodeller
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, 08034, Barcelona, Spain
| | - Ain Uustare
- ToxInvent LLC, Tiigi 61b, 50410, Tartu, Estonia
| | | | - Andrus Tasa
- ToxInvent LLC, Tiigi 61b, 50410, Tartu, Estonia
| | | | - Liis Salumäe
- Department, of Pathology, Tartu University Hospital, 50410, Tartu, Estonia
| | - Kazuki N Sugahara
- Division of GI/Endocrine Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lorena Simón-Gracia
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, 50411, Tartu, Estonia.
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, 50411, Tartu, Estonia.
- Materials Research Laboratory, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
8
|
Xiao X, Zheng Y, Wang T, Zhang X, Fang G, Zhang Z, Zhang Z, Zhao J. Enhancing anti-angiogenic immunotherapy for melanoma through injectable metal-organic framework hydrogel co-delivery of combretastatin A4 and poly(I:C). NANOSCALE ADVANCES 2024; 6:3135-3145. [PMID: 38868828 PMCID: PMC11166098 DOI: 10.1039/d4na00079j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
The interplay between vascularization and macrophage-induced immune suppression plays a crucial role in melanoma treatment. In this study, we propose a novel combination approach to combat melanoma by simultaneously inhibiting tumor vascularization and enhancing macrophage-mediated anti-tumor responses. We investigate the potential of combining combretastatin A4 (CA4), a vascular-disrupting agent, with poly(I:C) (PIC), an immunostimulatory adjuvant. This combination approach effectively suppresses melanoma cell proliferation, disrupts vascularization, and promotes macrophage polarization towards the M1 phenotype for melanoma suppression. To facilitate efficient co-delivery of CA4 and PIC for enhanced anti-angiogenic immunotherapy, we develop an injectable metal-organic framework hydrogel using Zeolitic Imidazolate Framework-8 (ZIF-8) and hyaluronic acid (HA) (ZIF-8/HA). Our findings demonstrate that ZIF-8 enables efficient loading of CA4 and enhances the stability of PIC against RNAase degradation in vitro. Furthermore, the developed co-delivery hydrogel system, PIC/CA4@ZIF-8/HA, exhibits improved rheological properties, good injectability and prolonged drug retention. Importantly, in vivo experiments demonstrate that the PIC/CA4@ZIF-8/HA formulation significantly reduces the dosage and administration frequency while achieving a more pronounced therapeutic effect. It effectively inhibits melanoma growth by suppressing angiogenesis, destroying blood vessels, promoting M1 macrophage infiltration, and demonstrating excellent biocompatibility. In conclusion, our study advances anti-angiogenic immunotherapy for melanoma through the potent combination of PIC/CA4, particularly when administered using the PIC/CA4@ZIF-8/HA formulation. These findings provide a new perspective on clinical anti-angiogenic immunotherapy for melanoma, emphasizing the importance of targeting tumor vascularization and macrophage-mediated immune suppression simultaneously.
Collapse
Affiliation(s)
- Xufeng Xiao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Yunuo Zheng
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital Xuzhou 221009 Jiangsu China
| | - Tianlong Wang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Xiaoqing Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Gaochuan Fang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University Xuzhou 221002 Jiangsu China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University Xuzhou 221002 Jiangsu China
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| |
Collapse
|
9
|
Czapla J, Drzyzga A, Ciepła J, Matuszczak S, Jarosz-Biej M, Pilny E, Cichoń T, Smolarczyk R. Combination of STING agonist with anti-vascular RGD-(KLAKLAK) 2 peptide as a novel anti-tumor therapy. Cancer Immunol Immunother 2024; 73:148. [PMID: 38832958 PMCID: PMC11150340 DOI: 10.1007/s00262-024-03732-3] [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: 04/05/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
Immunotherapy is one of the most promising anti-cancer treatment. It involves activating the host's own immune system to eliminate cancer cells. Activation of cGAS-STING pathway is promising therapeutic approach for cancer immunotherapy. However, in human clinical trials, targeting cGAS-STING pathway results in insufficient or unsustainable anti-tumor response. To enhance its effectiveness, combination with other anti-cancer therapies seems essential to achieve synergistic systemic anti-tumor response.The aim of this study was to evaluate whether the combination of STING agonist-cGAMP with anti-vascular RGD-(KLAKLAK)2 peptide results in a better anti-tumor response in poorly immunogenic tumors with various STING protein and αvβ3 integrin status.Combination therapy inhibited growth of murine breast carcinoma more effectively than melanoma. In melanoma, the administration of STING agonist alone was sufficient to obtain a satisfactory therapeutic effect. In both tumor models we have noted stimulation of innate immune response following cGAMP administration alone or in combination. The largest population of immune cells infiltrating the TME after therapy were activated NK cells. Increased infiltration of cytotoxic CD8+ T lymphocytes within the TME was only observed in melanoma tumors. However, they also expressed the "exhaustion" PD-1 receptor. In contrast, in breast carcinoma tumors each therapy caused the drop in the number of infiltrating CD8+ T cells.The obtained results indicate an additional therapeutic benefit from combining STING agonist with an anti-vascular agent. However, this effect depends on the type of tumor, the status of its microenvironment and the expression of specific proteins such as STING and αvβ3 family integrin.
Collapse
Affiliation(s)
- Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.
| | - Alina Drzyzga
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Joanna Ciepła
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Ewelina Pilny
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.
| |
Collapse
|
10
|
Izmest'ev AN, Svirshchevskaya EV, Akopov SB, Kravchenko AN, Gazieva GA. Recognition of arylmethylidene derivatives of imidazothiazolotriazinones as novel tubulin polymerization inhibitors. RSC Med Chem 2024; 15:1258-1273. [PMID: 38665841 PMCID: PMC11042243 DOI: 10.1039/d4md00027g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/25/2024] [Indexed: 04/28/2024] Open
Abstract
Two series of arylmethylidene derivatives of imidazothiazolotriazinone differing in the structure of the imidazothiazolotriazine fragment were synthesized and their antiproliferative activity and effect on tubulin polymerization were evaluated. Some of the synthesized derivatives showed a significant antiproliferative effect, among which (Z)-7-(2,4-dichlorobenzylidene)-1,3-diethyl-1,3a,4,9a-tetrahydroimidazo[4,5-e]thiazolo[2,3-c][1,2,4]triazine-2,8(3H,7H)-dione 2n exhibited the highest antiproliferative activity. The GI50 values of the compound against 56 of the 58 cell lines were 19.4-87.8 nM; against the remaining 2 cell lines, they were 0.544-1.29 μM. Moreover, further mechanism analysis demonstrated that 2n caused G2/M arrest, induced cell apoptosis in K562 cells and blocked tubulin polymerization in the same way as colchicine.
Collapse
Affiliation(s)
- Alexei N Izmest'ev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Moscow 119991 Russian Federation
| | - Elena V Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences Moscow 117997 Russian Federation
| | - Sergey B Akopov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences Moscow 117997 Russian Federation
| | - Angelina N Kravchenko
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Moscow 119991 Russian Federation
| | - Galina A Gazieva
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Moscow 119991 Russian Federation
| |
Collapse
|
11
|
Matsumoto K, Sunaga Y, Ecstein-Fraisse E, Fujiwara K. Phase I study of ombrabulin in combination with paclitaxel and carboplatin in Japanese patients with advanced solid tumors. Int J Gynecol Cancer 2024; 34:586-593. [PMID: 37989482 DOI: 10.1136/ijgc-2022-003880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVES To evaluate the maximum tolerated dose/maximum administered dose, safety, pharmacokinetic, and efficacy profiles of ombrabulin combined with paclitaxel and carboplatin in Japanese patients with solid tumors. METHODS Ombrabulin (25, 30, or 35 mg/m2) combined with paclitaxel (175 or 200 mg/m2) and carboplatin (AUC5 or AUC6) was administered by intravenous infusion once every 3 weeks to patients with advanced solid tumors, including cervical, ovarian, and uterine cancers. The maximum tolerated dose/maximum administered dose was defined based on the dose-limiting toxicity at cycle 1. Efficacy was assessed based on Response Evaluation Criteria In Solid Tumors (RECIST). RESULTS In total, 18 patients were recruited for this dose escalation study. One out of six patients treated with the highest doses of combination of ombrabulin (35 mg/m2), paclitaxel (200 mg/m2), and carboplatin (AUC6) presented a dose-limiting toxicity consisting of grade 3 Escherichia urinary tract infection. This dose was defined as the maximum tolerated dose of ombrabulin. The most frequent treatment-emergent adverse events were alopecia (83.3%), neutropenia and fatigue (72.2% each), decreased appetite, nausea, diarrhea, arthralgia, and myalgia (66.7% each). The grade 3-4 treatment-emergent adverse events included neutropenia (61.1%), Escherichia urinary tract infection, drug hypersensitivity, syncope, pulmonary embolism, and hydronephrosis (one patient each). In efficacy evaluation, seven patients achieved partial response or better (38.9%), including one complete response, and seven of 18 patients had stable disease (38.9%). Pharmacokinetic profiles in this Japanese study were comparable with those observed in the previous study without Japanese patients. CONCLUSIONS Although the maximum tolerated dose/maximum administered dose of ombrabulin (35 mg/m2) with taxane-platinum combination may be tolerable in Japanese patients in the first cycle, the dosages in the repeated treatment should be carefully selected for further study. TRIAL REGISTRATION NUMBER NCT01293630.
Collapse
Affiliation(s)
- Koji Matsumoto
- Division of Medical Oncology, Hyogo Cancer Center, Akashi, Hyogo, Japan
| | | | | | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| |
Collapse
|
12
|
Zalatan JG, Petrini L, Geiger R. Engineering bacteria for cancer immunotherapy. Curr Opin Biotechnol 2024; 85:103061. [PMID: 38219524 PMCID: PMC10922846 DOI: 10.1016/j.copbio.2023.103061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/30/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
Bacterial therapeutics have emerged as promising delivery systems to target tumors. These engineered live therapeutics can be harnessed to modulate the tumor microenvironment or to deliver and selectively release therapeutic payloads to tumors. A major challenge is to deliver bacteria systemically without causing widespread inflammation, which is critical for the many tumors that are not accessible to direct intratumoral injection. We describe potential strategies to address this challenge, along with approaches for specific payload delivery and biocontainment to ensure safety. These strategies will pave the way for the development of cost-effective, widely applicable next-generation cancer therapeutics.
Collapse
Affiliation(s)
- Jesse G Zalatan
- Department of Chemistry, University of Washington, Seattle, WA, United States.
| | - Lorenzo Petrini
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland; Institute of Oncology Research, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
13
|
Basu D, Dastidar SG. Molecular Dynamics and Machine Learning reveal distinguishing mechanisms of Competitive Ligands to perturb α,β-Tubulin. Comput Biol Chem 2024; 108:108004. [PMID: 38157659 DOI: 10.1016/j.compbiolchem.2023.108004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/25/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024]
Abstract
The mechanisms of action of ligands competing for the Colchicine Binding Site (CBS) of the α,β-Tubulin are non-standard compared to the commonly witnessed ligand-induced inhibition of proteins. This is because their potencies are not solely judged by the binding affinity itself, but also by their capacity to bias the conformational states of the dimer. Regarding the latter requirement, it is observed that ligands competing for the same pocket that binds colchicine exhibit divergence in potential clinical outcomes. Molecular dynamics-based ∼5.2 µs sampling of α,β-Tubulin complexed with four different ligands has revealed that each ligand has its customized way of influencing the complex. Primarily, it is the proportion of twisting and/or bending characteristic of modes of the intrinsic dynamics which is revealed to be 'fundamental' to tune this variation in the mechanism. The milder influence of 'bending' makes a ligand (TUB092), better classifiable under the group of vascular disrupting agents (VDAs), which are phenotypically effective on cytoskeletons; whereas a stronger impact of 'bending' makes the classical ligand Colchicine (COL) a better Anti-Mitotic agent (AMA). Two other ligands BAL27862 (2RR) and Nocodazole (NZO) fall in the intermediate zone as they fail to explicitly induce bending modes. Random Forest Classification method and K-means Clustering is applied to reveal the efficiency of Machine Learning methods in classifying the Tubulin conformations according to their ligand-specific perturbations and to highlight the significance of specific amino acid residues, mostly positioned in the α-β and β-β interfaces involved in the mechanism. These key residues responsible to yield discriminative actions of the ligands are likely to be highly useful in future endeavours to design more precise inhibitors.
Collapse
Affiliation(s)
- Debadrita Basu
- Biological Sciences, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Shubhra Ghosh Dastidar
- Biological Sciences, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India.
| |
Collapse
|
14
|
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
|
15
|
Bian S, Zheng X, Liu W, Gao Z, Wan Y, Li J, Ren H, Zhang W, Lee CS, Wang P. pH-Responsive NIR-II phototheranostic agents for in situ tumor vascular monitoring and combined anti-vascular/photothermal therapy. Biomaterials 2023; 303:122380. [PMID: 37925793 DOI: 10.1016/j.biomaterials.2023.122380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
Developing nanoplatforms integrating superior fluorescence imaging ability in second near-infrared (NIR-II) window and tumor microenvironment responsive multi-modal therapy holds great potential for real-time feedback of therapeutic efficacy and optimizing tumor inhibition. Herein, we developed a pH-sensitive pyrrolopyrrole aza-BODIPY-based amphiphilic molecule (PTG), which has a balanced NIR-II fluorescence brightness and photothermal effect. PTG is further co-assembled with a vascular disrupting agent (known as DMXAA) to prepare PTDG nanoparticles for combined anti-vascular/photothermal therapy and real-time monitoring of the tumor vascular disruption. Each PTG molecule has an active PT-3 core which is linked to two PEG chains via pH-sensitive ester bonds. The cleavage of ester bonds in the acidic tumor environment would tricker releases of DMXAA for anti-vascular therapy and further assemble PT-3 cores into micrometer particles for long term monitoring of the tumor progression. Furthermore, benefiting from the high brightness in the NIR-II region (119.61 M-1 cm-1) and long blood circulation time (t1/2 = 235.6 min) of PTDG nanoparticles, the tumor vascular disrupting process can be in situ visualized in real time during treatment. Overall, this study demonstrates a self-assembly strategy to build a pH-responsive NIR-II nanoplatform for real-time monitoring of tumor vascular disruption, long-term tracking tumor progression and combined anti-vascular/photothermal therapy.
Collapse
Affiliation(s)
- Shuaishuai Bian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiuli Zheng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Weimin Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zekun Gao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingpeng Wan
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Jihao Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haohui Ren
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wenjun Zhang
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Chun-Sing Lee
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China.
| | - Pengfei Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
16
|
Chen L, Hu Y, Lu Z, Lin Z, Li L, Wu JQ, Yu ZL, Wang C, Chen WH, Hu J. Design, Synthesis, and Antitumor Efficacy of Substituted 2-Amino[1,2,4]triazolopyrimidines and Related Heterocycles as Dual Inhibitors for Microtubule Polymerization and Janus Kinase 2. J Med Chem 2023; 66:15006-15024. [PMID: 37856840 DOI: 10.1021/acs.jmedchem.3c01690] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Preclinical and clinical studies have demonstrated the synergistic effect of microtubule-targeting agents in combination with Janus kinase 2 (JAK2) inhibitors, prompting the development of single agents with enhanced therapeutic efficacy by dually inhibiting tubulin polymerization and JAK2. Herein, we designed and synthesized a series of substituted 2-amino[1,2,4]triazolopyrimidines and related heterocycles as dual inhibitors for tubulin polymerization and JAK2. Most of these compounds exhibited potent antiproliferative activity against the selected cancer cells, with compound 7g being the most active. This compound effectively inhibits both tubulin assembly and JAK2 activity. Furthermore, phosphorylated compound 7g (i.e., compound 7g-P) could efficiently convert to compound 7g in vivo. Compound 7g, whether it was administered directly or in the form of a phosphorylated prodrug (i.e., compound 7g-P), significantly inhibited the growth of A549 xenografts in nude mice. The present findings strongly suggest that compound 7g represents a promising chemotherapeutic agent with high antitumor efficacy.
Collapse
Affiliation(s)
- Li Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Yunfei Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Zhonghui Lu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Zeyin Lin
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Lanqing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Jia-Qiang Wu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Zhi-Ling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 852, P. R. China
| | - Chunye Wang
- Department of Pharmacy, Huizhou First Maternal and Child Health Care Hospital, Huizhou 516000, P. R. China
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| | - Jinhui Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, P. R. China
| |
Collapse
|
17
|
Wang C, Xu J, Zhang Y, Nie G. Emerging nanotechnological approaches to regulating tumor vasculature for cancer therapy. J Control Release 2023; 362:647-666. [PMID: 37703928 DOI: 10.1016/j.jconrel.2023.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Abnormal angiogenesis stands for one of the most striking manifestations of malignant tumor. The pathologically and structurally abnormal tumor vasculature facilitates a hostile tumor microenvironment, providing an ideal refuge exclusively for cancer cells. The emergence of vascular regulation drugs has introduced a distinctive class of therapeutics capable of influencing nutrition supply and drug delivery efficacy without the need to penetrate a series of physical barriers to reach tumor cells. Nanomedicines have been further developed for more precise regulation of tumor vasculature with the capacity of co-delivering multiple active pharmaceutical ingredients, which overall reduces the systemic toxicity and boosts the therapeutic efficacy of free drugs. Additionally, precise structure design enables the integration of specific functional motifs, such as surface-targeting ligands, droppable shells, degradable framework, or stimuli-responsive components into nanomedicines, which can improve tissue-specific accumulation, enhance tissue penetration, and realize the controlled and stimulus-triggered release of the loaded cargo. This review describes the morphological and functional characteristics of tumor blood vessels and summarizes the pivotal molecular targets commonly used in nanomedicine design, and then highlights the recent cutting-edge advancements utilizing nanotechnologies for precise regulation of tumor vasculature. Finally, the challenges and future directions of this field are discussed.
Collapse
Affiliation(s)
- Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yinlong Zhang
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; School of Nanoscience and Engineering, School of Chemical Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China.
| |
Collapse
|
18
|
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
|
19
|
Lahooti B, Akwii RG, Zahra FT, Sajib MS, Lamprou M, Alobaida A, Lionakis MS, Mattheolabakis G, Mikelis CM. Targeting endothelial permeability in the EPR effect. J Control Release 2023; 361:212-235. [PMID: 37517543 DOI: 10.1016/j.jconrel.2023.07.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
The characteristics of the primary tumor blood vessels and the tumor microenvironment drive the enhanced permeability and retention (EPR) effect, which confers an advantage towards enhanced delivery of anti-cancer nanomedicine and has shown beneficial effects in preclinical models. Increased vascular permeability is a landmark feature of the tumor vessels and an important driver of the EPR. The main focus of this review is the endothelial regulation of vascular permeability. We discuss current challenges of targeting vascular permeability towards clinical translation and summarize the structural components and mechanisms of endothelial permeability, the principal mediators and signaling players, the targeted approaches that have been used and their outcomes to date. We also critically discuss the effects of the tumor-infiltrating immune cells, their interplay with the tumor vessels and the impact of immune responses on nanomedicine delivery, the impact of anti-angiogenic and tumor-stroma targeting approaches, and desirable nanoparticle design approaches for greater translational benefit.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Margarita Lamprou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece.
| |
Collapse
|
20
|
Paresishvili T, Kakabadze Z. Challenges and Opportunities Associated With Drug Delivery for the Treatment of Solid Tumors. Oncol Rev 2023; 17:10577. [PMID: 37711860 PMCID: PMC10497757 DOI: 10.3389/or.2023.10577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/17/2023] [Indexed: 09/16/2023] Open
Abstract
In this review, we discuss the effectiveness of drug delivery system based on metal nanoparticles, and also, describe the problems associated with their delivery to tumor cells. Throughout recent years, more reports have appeared in the literature that demonstrate promising results for the treatment of various types of cancer using metal-based nanoparticles. Due to their unique physical and chemical properties, metal nanoparticles are effectively being used for the delivery of drug to the tumor cells, for cancer diagnosis and treatment. They can also be synthesized allowing the control of size and shape. However, the effectiveness of the metal nanoparticles for cancer treatment largely depends on their stability, biocompatibility, and ability to selectively affect tumor cells after their systemic or local administration. Another major problem associated with metal nanoparticles is their ability to overcome tumor tissue barriers such as atypical blood vessel structure, dense and rigid extracellular matrix, and high pressure of tumor interstitial fluid. The review also describes the design of tumor drug delivery systems that are based on metal nanoparticles. The mechanism of action of metal nanoparticles on cancer cells is also discussed. Considering the therapeutic safety and toxicity of metal nanoparticles, the prospects for their use for future clinical applications are being currently reviewed.
Collapse
Affiliation(s)
- Teona Paresishvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | | |
Collapse
|
21
|
Czapla J, Drzyzga A, Matuszczak S, Cichoń T, Rusin M, Jarosz-Biej M, Pilny E, Smolarczyk R. Antitumor effect of anti-vascular therapy with STING agonist depends on the tumor microenvironment context. Front Oncol 2023; 13:1249524. [PMID: 37655095 PMCID: PMC10465696 DOI: 10.3389/fonc.2023.1249524] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
Introduction Targeting tumor vasculature is an efficient weapon to fight against cancer; however, activation of alternative pathways to rebuild the disrupted vasculature leads to rapid tumor regrowth. Immunotherapy that exploits host immune cells to elicit and sustain potent antitumor response has emerged as one of the most promising tools for cancer treatment, yet many treatments fail due to developed resistance mechanisms. Therefore, our aim was to examine whether combination of immunotherapy and anti-vascular treatment will succeed in poorly immunogenic, difficult-to-treat melanoma and triple-negative breast tumor models. Methods Our study was performed on B16-F10 melanoma and 4T1 breast tumor murine models. Mice were treated with the stimulator of interferon genes (STING) pathway agonist (cGAMP) and vascular disrupting agent combretastatin A4 phosphate (CA4P). Tumor growth was monitored. The tumor microenvironment (TME) was comprehensively investigated using multiplex immunofluorescence and flow cytometry. We also examined if such designed therapy sensitizes investigated tumor models to an immune checkpoint inhibitor (anti-PD-1). Results The use of STING agonist cGAMP as monotherapy was insufficient to effectively inhibit tumor growth due to low levels of STING protein in 4T1 tumors. However, when additionally combined with an anti-vascular agent, a significant therapeutic effect was obtained. In this model, the obtained effect was related to the TME polarization and the stimulation of the innate immune response, especially activation of NK cells. Combination therapy was unable to activate CD8+ T cells. Due to the lack of PD-1 upregulation, no improved therapeutic effect was observed when additionally combined with the anti-PD-1 inhibitor. In B16-F10 tumors, highly abundant in STING protein, cGAMP as monotherapy was sufficient to induce potent antitumor response. In this model, the therapeutic effect was due to the infiltration of the TME with activated NK cells. cGAMP also caused the infiltration of CD8+PD-1+ T cells into the TME; hence, additional benefits of using the PD-1 inhibitor were observed. Conclusion The study provides preclinical evidence for a great influence of the TME on the outcome of applied therapy, including immune cell contribution and ICI responsiveness. We pointed the need of careful TME screening prior to antitumor treatments to achieve satisfactory results.
Collapse
Affiliation(s)
- Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | | | | | | | | | | | | | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| |
Collapse
|
22
|
Xiong Y, Rao Y, Hu J, Luo Z, Chen C. Nanoparticle-Based Photothermal Therapy for Breast Cancer Noninvasive Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2305140. [PMID: 37561994 DOI: 10.1002/adma.202305140] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/29/2023] [Indexed: 08/12/2023]
Abstract
Rapid advancements in materials science and nanotechnology, intertwined with oncology, have positioned photothermal therapy (PTT) as a promising noninvasive treatment strategy for cancer. The breast's superficial anatomical location and aesthetic significance render breast cancer a particularly pertinent candidate for the clinical application of PTT following melanoma. This review comprehensively explores the research conducted on the various types of nanoparticles employed in PTT for breast cancer and elaborates on their specific roles and mechanisms of action. The integration of PTT with existing clinical therapies for breast cancer is scrutinized, underscoring its potential for synergistic outcomes. Additionally, the mechanisms underlying PTT and consequential modifications to the tumor microenvironment after treatment are elaborated from a medical perspective. Future research directions are suggested, with an emphasis on the development of integrative platforms that combine multiple therapeutic approaches and the optimization of nanoparticle synthesis for enhanced treatment efficacy. The goal is to push the boundaries of PTT toward a comprehensive, clinically applicable treatment for breast cancer.
Collapse
Affiliation(s)
- Yao Xiong
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Yan Rao
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, Hubei, 430000, P. R. China
| | - Jiawei Hu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Zixuan Luo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| |
Collapse
|
23
|
Nagashima Y, Tochinai R, Sekizawa SI, Kato D, Nakagawa T, Tsuru Y, Tatewaki Y, Mutoh T, Taki Y, Kuwahara M. Pretreatment with tadalafil attenuates cardiotoxicity induced by combretastatin A4 disodium phosphate in rats. J Toxicol Pathol 2023; 36:151-158. [PMID: 37577366 PMCID: PMC10412959 DOI: 10.1293/tox.2022-0143] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/04/2023] [Indexed: 08/15/2023] Open
Abstract
Combretastatin A4 disodium phosphate (CA4DP) is a prodrug of combretastatin A4 (CA4), a microtubule-disassembling agent that exhibits antitumor effects by inhibiting tumor cell proliferation and inducing morphological changes and apoptosis in vascular endothelial cells in tumors. However, cardiotoxicity induced by ischemia and hypertension is a severe adverse event. In this study, we focused on the fact that phosphodiesterase (PDE) 5 inhibitors dilate the heart and peripheral blood vessels and aimed to investigate whether co-administration of tadalafil, a PDE5 inhibitor, can attenuate cardiotoxicity without altering the antitumor effect of CA4DP. To investigate cardiotoxicity, CA4DP and/or tadalafil were administered to rats, and blood pressure, echocardiography, histopathology, and cGMP concentration in the myocardium were examined. Administration of CA4DP increased systolic blood pressure, decreased cardiac function, lowered cGMP levels in the myocardium, and led to necrosis of myocardial cells. Co-administration of tadalafil attenuated these CA4DP-induced changes. To investigate the antitumor effect, canine mammary carcinoma cell lines (CHMp-13a) and human umbilical vein endothelial cells were cultured with CA4 and/or tadalafil, and cell proliferation and endothelial vascular tube disruption were examined. CHMp-13a cells were transplanted into nude mice and treated with CA4DP and/or tadalafil. CA4-induced inhibition of cell proliferation and disruption of the endothelial vascular tube were not affected by co-treatment with tadalafil, and the antitumor effects of CA4DP in xenograft mice were not reduced by co-administration of tadalafil. These results revealed that myocardial damage induced by CA4DP was attenuated by co-administration of tadalafil while maintaining antitumor efficacy.
Collapse
Affiliation(s)
- Yoshiyasu Nagashima
- Department of Veterinary Pathophysiology and Animal Health,
Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi,
Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryota Tochinai
- Department of Veterinary Pathophysiology and Animal Health,
Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi,
Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Aging Research and Geriatric Medicine,
Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryocho, Aobaku,
Sendai 980-8575, Japan
- Research Institute for Brain and Blood Vessels, Akita
Cerebrospinal and Cardiovascular Center, 6-10 Sensyu-Kubota-machi, Akita 010-0874,
Japan
| | - Shin-ichi Sekizawa
- Department of Veterinary Pathophysiology and Animal Health,
Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi,
Bunkyo-ku, Tokyo 113-8657, Japan
| | - Daiki Kato
- Laboratory of Veterinary Surgery, Graduate School of
Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo,
113-8657, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of
Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo,
113-8657, Japan
| | - Yoshiharu Tsuru
- Primetech Corp. Life Science Laboratory, 1-1-1 Yayoi,
Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Yasuko Tatewaki
- Department of Aging Research and Geriatric Medicine,
Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryocho, Aobaku,
Sendai 980-8575, Japan
| | - Tatsushi Mutoh
- Department of Aging Research and Geriatric Medicine,
Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryocho, Aobaku,
Sendai 980-8575, Japan
- Research Institute for Brain and Blood Vessels, Akita
Cerebrospinal and Cardiovascular Center, 6-10 Sensyu-Kubota-machi, Akita 010-0874,
Japan
| | - Yasuyuki Taki
- Department of Aging Research and Geriatric Medicine,
Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryocho, Aobaku,
Sendai 980-8575, Japan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health,
Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi,
Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
24
|
Hautanen V, Toimela T, Paparella M, Heinonen T. A Human Cell-based Assay to Assess the Induction of Vasculature Formation for Non-genotoxic Carcinogenicity Testing Purposes: A Pilot Study. Altern Lab Anim 2023:2611929231171165. [PMID: 37125451 DOI: 10.1177/02611929231171165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The induction of vasculature formation is proposed to be a significant mechanism behind the non-genotoxic carcinogenicity of a chemical. The vasculature formation model used in this study is based on the coculture of human primary HUVECs and hASCs. This model was used to develop an assay to assess the induction of vasculature formation. Three assay protocols, based on different conditions, were developed and compared in order to identify the optimal conditions required. Some serum supplements and growth factors were observed to be essential for initiating vasculature formation. Of the studied putative positive reference chemicals, aspartame, sodium nitrite, bisphenol A and nicotine treatment led to a clear induction of vasculature formation, but arsenic and cadmium treatment only led to a slight increase. This human cell-based assay has the potential to be used as one test within a next generation testing battery, to assess the non-genotoxic carcinogenicity of a chemical through the mechanism of vasculature formation induction.
Collapse
Affiliation(s)
- Veera Hautanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tarja Toimela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Martin Paparella
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Tuula Heinonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
25
|
Ma X, Fang W, Wang D, Shao N, Chen J, Nie T, Huang C, Huang Y, Luo L, Xiao Z. Nanomaterial-Based Antivascular Therapy in the Multimodal Treatment of Cancer. Pharmaceutics 2023; 15:pharmaceutics15041207. [PMID: 37111692 PMCID: PMC10145863 DOI: 10.3390/pharmaceutics15041207] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Abnormal tumor vasculature and a hypoxic tumor microenvironment (TME) limit the effectiveness of conventional cancer treatment. Recent studies have shown that antivascular strategies that focus on antagonizing the hypoxic TME and promoting vessel normalization effectively synergize to increase the antitumor efficacy of conventional therapeutic regimens. By integrating multiple therapeutic agents, well-designed nanomaterials exhibit great advantages in achieving higher drug delivery efficiency and can be used as multimodal therapy with reduced systemic toxicity. In this review, strategies for the nanomaterial-based administration of antivascular therapy combined with other common tumor treatments, including immunotherapy, chemotherapy, phototherapy, radiotherapy, and interventional therapy, are summarized. In particular, the administration of intravascular therapy and other therapies with the use of versatile nanodrugs is also described. This review provides a reference for the development of multifunctional nanotheranostic platforms for effective antivascular therapy in combined anticancer treatments.
Collapse
Affiliation(s)
- Xiaocong Ma
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Weimin Fang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Tianqi Nie
- The 12th People's Hospital of Guangzhou, Guangzhou 510620, China
| | - Cuiqing Huang
- Department of Ultrasound, Guangdong Women and Children Hospital, Guangzhou 511400, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|
26
|
Jin Q, Chen D, Song Y, Liu T, Li W, Chen Y, Qin X, Zhang L, Wang J, Xie M. Ultrasound-Responsive Biomimetic Superhydrophobic Drug-Loaded Mesoporous Silica Nanoparticles for Treating Prostate Tumor. Pharmaceutics 2023; 15:pharmaceutics15041155. [PMID: 37111641 PMCID: PMC10146986 DOI: 10.3390/pharmaceutics15041155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/26/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023] Open
Abstract
Interfacial nanobubbles on a superhydrophobic surface can serve as ultrasound cavitation nuclei for continuously promoting sonodynamic therapy, but their poor dispersibility in blood has limited their biomedical application. In this study, we proposed ultrasound-responsive biomimetic superhydrophobic mesoporous silica nanoparticles, modified with red blood cell membrane and loaded with doxorubicin (DOX) (F-MSN-DOX@RBC), for RM-1 tumor sonodynamic therapy. Their mean size and zeta potentials were 232 ± 78.8 nm and −35.57 ± 0.74 mV, respectively. The F-MSN-DOX@RBC accumulation in a tumor was significantly higher than in the control group, and the spleen uptake of F-MSN-DOX@RBC was significantly reduced in comparison to that of the F-MSN-DOX group. Moreover, the cavitation caused by a single dose of F-MSN-DOX@RBC combined with multiple ultrasounds provided continuous sonodynamic therapy. The tumor inhibition rates in the experimental group were 71.5 8 ± 9.54%, which is significantly better than the control group. DHE and CD31 fluorescence staining was used to assess the reactive oxygen species (ROS) generated and the broken tumor vascular system induced by ultrasound. Finally, we can conclude that the combination of anti-vascular therapy, sonodynamic therapy by ROS, and chemotherapy promoted tumor treatment efficacy. The use of red blood cell membrane-modified superhydrophobic silica nanoparticles is a promising strategy in designing ultrasound-responsive nanoparticles to promote drug-release.
Collapse
Affiliation(s)
- Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dandan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yishu Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Tianshu Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
27
|
Liu ZL, Ren XT, Huang Y, Sun JL, Wang XS, Zheng MF, Cui LJ, Zhang XF, Tang ZH. A Novel CA4P Polymeric Nanoparticle for Murine Hepatoma Therapy. CHINESE JOURNAL OF POLYMER SCIENCE 2023. [DOI: 10.1007/s10118-023-2921-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
28
|
Tumor vasculature VS tumor cell targeting: Understanding the latest trends in using functional nanoparticles for cancer treatment. OPENNANO 2023. [DOI: 10.1016/j.onano.2023.100136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
29
|
Li Y, Zhang J, Lu C, Guo M, Zhang J, Huang G, Ni Y, Chen Y. Feasibility of the novel vascular disrupting agent C118P for facilitating high-intensity focused ultrasound ablation of uterine fibroids. Int J Hyperthermia 2023; 40:2185576. [PMID: 36913972 DOI: 10.1080/02656736.2023.2185576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
OBJECTIVE In this study, C118P, a novel vascular disrupting agent (VDA), was evaluated for its ability in improving the ablative effect of high-intensity focused ultrasound (HIFU) on uterine fibroids by reducing blood perfusion. METHODS Eighteen female rabbits were infused with isotonic sodium chloride solution (ISCS), C118P or oxytocin for 30 min, and an HIFU ablation of the leg muscles was performed within the last 2 min. Blood pressure, heart rate and laser speckle flow imaging (LSFI) of the auricular blood vessels were recorded during perfusion. Ears with vessels, uterus and muscle ablation sites were collected and sliced for hematoxylin-eosin (HE) staining to compare vascular size, as well as nicotinamide adenine dinucleotide-tetrazolium reductase (NADH-TR) staining to observe necrosis after ablation. RESULTS Analyses revealed that the perfusion of C118P or oxytocin steadily reduced blood perfusion in the ears to approximately half by the end of the perfusion, constricted the blood vessels of the ears and uterus, and improved HIFU ablation in the muscle tissues. C118P increased blood pressure and decreased heart rate. The degree of contraction of the auricular and uterine blood vessels was positively correlated. CONCLUSION This study confirmed that C118P could reduce blood perfusion in various tissues and had a better synergistic effect with HIFU ablation of muscle (the same tissue type as fibroids) than did oxytocin. C118P could therefore possibly replace oxytocin in facilitating HIFU ablation of uterine fibroids; however, electrocardiographic monitoring is required.
Collapse
Affiliation(s)
- Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jinyong Zhang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Changlian Lu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Mingrui Guo
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jue Zhang
- College of Medical Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yicheng Ni
- Department of Imaging & Pathology, Biomedical Sciences Group, Leuven, Belgium
| | - Yini Chen
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicinel, Shanghai, China.,Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| |
Collapse
|
30
|
Kim SJ, Lee K, Park J, Park M, Kim UJ, Kim SM, Ryu KH, Kang KW. CKD-516 potentiates the anti-cancer activity of docetaxel against epidermal growth factor receptor tyrosine kinase inhibitor-resistant lung cancer. Toxicol Res 2023; 39:61-69. [PMID: 36726834 PMCID: PMC9839922 DOI: 10.1007/s43188-022-00146-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Lung cancer is the leading cause of cancer death. Although docetaxel has been used as a second- or third-line treatment for non-small cell lung cancer (NSCLC), the objective response rate is less than 10%. Hence, there is a need to improve the clinical efficacy of docetaxel monotherapy; combination therapy should be considered. Here, we show that CKD-516, a vascular disruption agent, can be combined with docetaxel to treat epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI)-resistant NSCLC. CKD-516 was orally bioavailable; neither CKD-516 nor docetaxel affected the mean plasma concentration-time profile or pharmacokinetic parameters of the other drug. CKD-516 and docetaxel synergistically inhibited the growth of H1975 (with an L858R/T790M double mutation of EGFR) and A549 (with a KRAS mutation) lung cancer cell lines. In addition, docetaxel plus CKD-516 delayed tumor growth in-and extended the lifespan of-tumor-bearing mice. Thus, combination CKD-516 and docetaxel therapy could be used to treat EGFR-TKI-resistant NSCLC.
Collapse
Affiliation(s)
- Soo Jin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826 Republic of Korea
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, 16995, Gyeonggi-do, Republic of Korea
| | - Kyunghyeon Lee
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, 16995, Gyeonggi-do, Republic of Korea
| | - Jaewoo Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826 Republic of Korea
| | - Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826 Republic of Korea
| | - U. Ji Kim
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, 16995, Gyeonggi-do, Republic of Korea
| | - Se-mi Kim
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, 16995, Gyeonggi-do, Republic of Korea
| | - Keun Ho Ryu
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, 16995, Gyeonggi-do, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826 Republic of Korea
| |
Collapse
|
31
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
32
|
Wu Z, Bian Y, Chu T, Wang Y, Man S, Song Y, Wang Z. The role of angiogenesis in melanoma: Clinical treatments and future expectations. Front Pharmacol 2022; 13:1028647. [PMID: 36588679 PMCID: PMC9797529 DOI: 10.3389/fphar.2022.1028647] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The incidence of melanoma has increased rapidly over the past few decades, with mortality accounting for more than 75% of all skin cancers. The high metastatic potential of Melanoma is an essential factor in its high mortality. Vascular angiogenic system has been proved to be crucial for the metastasis of melanoma. An in-depth understanding of angiogenesis will be of great benefit to melanoma treatment and may promote the development of melanoma therapies. This review summarizes the recent advances and challenges of anti-angiogenic agents, including monoclonal antibodies, tyrosine kinase inhibitors, human recombinant Endostatin, and traditional Chinese herbal medicine. We hope to provide a better understanding of the mechanisms, clinical research progress, and future research directions of melanoma.
Collapse
Affiliation(s)
- Zhuzhu Wu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yifei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuman Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Yongmei Song
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Zhenguo Wang
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| |
Collapse
|
33
|
Faria P, Pacheco C, Moura RP, Sarmento B, Martins C. Multifunctional nanomedicine strategies to manage brain diseases. Drug Deliv Transl Res 2022; 13:1322-1342. [PMID: 36344871 DOI: 10.1007/s13346-022-01256-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Brain diseases represent a substantial social and economic burden, currently affecting one in six individuals worldwide. Brain research has been focus of great attention in order to unravel the pathogenesis and complexity of brain diseases at the cellular, molecular, and microenvironmental levels. Due to the intrinsic nature of the brain, the presence of the highly restrictive blood-brain barrier (BBB), and the pathophysiology of most diseases, therapies can hardly be considered successful purely by the administration of one drug to a patient. Apart from improving pharmacokinetic parameters, tailoring biodistribution, and reducing the number of side effects, nanomedicines are able to actively co-target the therapeutics to the brain parenchyma and brain lesions, as well as to achieve the delivery of multiple cargos with therapeutic, diagnostic, and theranostic properties. Among other multivalent effects that can be personalized according to the disease needs, this represents a promising class of novel nanosystems, termed multifunctional nanomedicines. Herein, we review the principal mechanisms of therapeutic resistance of the most prevalent brain diseases, how to overcome this therapeutic resistance through the use of multifunctional nanomedicines that tackle multiple fronts of the disease microenvironment, and the promising therapeutic responses achieved by some of the most cutting-edge multifunctional nanomedicines reported in literature.
Collapse
|
34
|
Guo K, Ma X, Li J, Zhang C, Wu L. Recent advances in combretastatin A-4 codrugs for cancer therapy. Eur J Med Chem 2022; 241:114660. [PMID: 35964428 DOI: 10.1016/j.ejmech.2022.114660] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
CA4 is a potent microtubule polymerization inhibitor and vascular disrupting agent. However, the in vivo efficiency of CA4 is limited owing to its poor pharmacokinetics resulting from its high lipophilicity and low water solubility. To improve the water solubility, CA4 phosphate (CA4P) has been developed and shows potent antivascular and antitumor effects. CA4P had been evaluated as a vascular disrupting agent in previousc linical trials. However, it had been discontinued due to the lack of a meaningful improvement in progression-free survival and unfavorable partial response data. Codrug is a drug design approach to chemically bind two or more drugs to improve therapeutic efficiency or decrease adverse effects. This review describes the progress made over the last twenty years in developing CA4-based codrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues. It also discusses the existing problems and the developmental prospects of CA4 codrugs.
Collapse
Affiliation(s)
- Kerong Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xin Ma
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jian Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
35
|
A self-activating nanoized vascular disrupting agent for selective anti-tumor therapy. Biomaterials 2022; 288:121736. [PMID: 35995623 DOI: 10.1016/j.biomaterials.2022.121736] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/23/2022] [Accepted: 08/06/2022] [Indexed: 12/29/2022]
Abstract
Vascular disrupting agents (VDAs) have great potential in antitumor therapy, while the efficiency is limited by cardiovascular toxicity. In this study, a self-activating nanoized plinabulin (poly (l-glutamic acid) grafted Azo-Plinabulin, AzoP-NP) was constructed. The AzoP-NPs can selectively be activated to an amino derivative of plinabulin (AmP) by intrinsic tumor hypoxia, disrupting tumor vessels and amplifying hypoxia, whilst be activated by self-amplified tumor hypoxia, then selectively inhibit tumor growth. In 4T1 tumor model, the AzoP-NPs had a selective biodistribution in tumor, as the free AmP in tumors at 24 h after AzoP-NPs treatment was 18.6 fold of that after AmP treatment and significantly higher than that in other tissues. Accordingly, AzoP-NPs resulted in no obvious acute cardiovascular toxicity (plasma von Willebrand factor in PBS, AzoP-NPs and AmP group: 143.1, 184.0 and 477.6 ng/mL) and a significantly stronger tumor inhibition than AmP. And the sustained release of drug in AzoP-NPs led to a higher maximum tolerated dose (MTD) (MTD of AzoP-NPs and AmP: > 80 vs 20 mg/kg). In addition, AzoP-NPs amplified tumor hypoxic, and synergized the anti-tumor effect of Tirapazamine (TPZ), a hypoxia-activated drug in clinical trials, with an inhibition rate of 97.7% and Q value of 1.89. Therefore, our findings provide new insights into next generation VDAs and their application in tumor therapy.
Collapse
|
36
|
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
|
37
|
Li F, Shao X, Liu D, Jiao X, Yang X, Yang W, Liu X. Vascular Disruptive Hydrogel Platform for Enhanced Chemotherapy and Anti-Angiogenesis through Alleviation of Immune Surveillance. Pharmaceutics 2022; 14:1809. [PMID: 36145556 PMCID: PMC9505154 DOI: 10.3390/pharmaceutics14091809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Patients undergoing immunotherapy always exhibit a low-response rate due to tumor heterogeneity and immune surveillance in the tumor. Angiogenesis plays an important role in affecting the status of tumor-infiltrated lymphocytes by inducing hypoxia and acidosis microenvironment, suggesting its synergistic potential in immunotherapy. However, the antitumor efficacy of singular anti-angiogenesis therapy often suffers from failure in the clinic due to the compensatory pro-angiogenesis signaling pathway. In this work, classic injectable thermosensitive PLGA-PEG-PLGA copolymer was used to construct a platform to co-deliver CA4P (vascular disruptive agent) and EPI for inducing immunogenic cell death of cancer cells by targeting the tumor immune microenvironment. Investigation of 4T1 tumor-bearing mouse models suggests that local administration of injectable V+E@Gel could significantly inhibit the proliferation of cancer cells and prolong the survival rate of 4T1 tumor-bearing mouse models. Histological analysis further indicates that V+E@Gel could effectively inhibit tumor angiogenesis and metastasis by down-regulating the expression of CD34, CD31, MTA1 and TGF-β. Moreover, due to the sustained release kinetics of V+E@Gel, its local administration relieves the immune surveillance in tumor tissues and thus induces a robust and long-lasting specific antitumor immune response. Overall, this work provides a new treatment strategy through the mediation of the tumor immune microenvironment by vascular disruption to fulfill enhanced chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Fasheng Li
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xinmei Shao
- Department of Neurology, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Dehui Liu
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xiaogang Jiao
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xinqi Yang
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Wencai Yang
- Department of Interventional, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xiaoyan Liu
- Department of Neurology, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| |
Collapse
|
38
|
Zhu H, Tan Y, He C, Liu Y, Duan Y, Zhu W, Zheng T, Li D, Xu J, Yang DH, Chen ZS, Xu S. Discovery of a Novel Vascular Disrupting Agent Inhibiting Tubulin Polymerization and HDACs with Potent Antitumor Effects. J Med Chem 2022; 65:11187-11213. [PMID: 35926141 DOI: 10.1021/acs.jmedchem.2c00681] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Most vascular disrupting agents (VDAs) fail to prevent the regrowth of blood vessels at the edge of tumors, causing tumor rebound and relapse. Herein, a series of novel multifunctional vascular disrupting agents (VDAs) capable of inhibiting microtubule polymerization and histone deacetylases (HDACs) were designed and synthesized using the tubulin polymerization inhibitor TH-0 as the lead compound. Among them, compound TH-6 exhibited the most potent antiproliferative activity (IC50 = 18-30 nM) against a panel of cancer cell lines. As expected, TH-6 inhibited tubulin assembly and increased the acetylation level of HDAC substrate proteins in HepG2 cells. Further in vivo antitumor assay displayed that TH-6 effectively inhibited tumor growth with no apparent toxicity. More importantly, TH-6 disrupted both the internal and peripheral tumor vasculatures, which contributed to the persistent tumor inhibitory effects after drug withdrawal. Altogether, TH-6 deserves to be further investigated for the new approach to clinical cancer therapy.
Collapse
Affiliation(s)
- Huajian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Yuchen Tan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Chen He
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Yang Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Yiping Duan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Wenjian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Tiandong Zheng
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, P. R. China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| |
Collapse
|
39
|
Zhang W, Yang L, Si W, Tang M, Bai P, Zhu Z, Kuang S, Liu J, Shi M, Huang J, Chen X, Li D, Wen Y, Yang Z, Xiao K, Chen L. SKLB-14b, a novel oral microtubule-destabilizing agent based on hydroxamic acid with potent anti-tumor and anti-multidrug resistance activities. Bioorg Chem 2022; 128:106053. [DOI: 10.1016/j.bioorg.2022.106053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 01/05/2023]
|
40
|
Yavvari P, Laporte A, Elomaa L, Schraufstetter F, Pacharzina I, Daberkow AD, Hoppensack A, Weinhart M. 3D-Cultured Vascular-Like Networks Enable Validation of Vascular Disruption Properties of Drugs In Vitro. Front Bioeng Biotechnol 2022; 10:888492. [PMID: 35769106 PMCID: PMC9234334 DOI: 10.3389/fbioe.2022.888492] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Vascular-disrupting agents are an interesting class of anticancer compounds because of their combined mode of action in preventing new blood vessel formation and disruption of already existing vasculature in the immediate microenvironment of solid tumors. The validation of vascular disruption properties of these drugs in vitro is rarely addressed due to the lack of proper in vitro angiogenesis models comprising mature and long-lived vascular-like networks. We herein report an indirect coculture model of human umbilical vein endothelial cells (HUVECs) and human dermal fibroblasts (HDFs) to form three-dimensional profuse vascular-like networks. HUVECs embedded and sandwiched in the collagen scaffold were cocultured with HDFs located outside the scaffold. The indirect coculture approach with the vascular endothelial growth factor (VEGF) producing HDFs triggered the formation of progressively maturing lumenized vascular-like networks of endothelial cells within less than 7 days, which have proven to be viably maintained in culture beyond day 21. Molecular weight-dependent Texas red-dextran permeability studies indicated high vascular barrier function of the generated networks. Their longevity allowed us to study the dose-dependent response upon treatment with the three known antiangiogenic and/or vascular disrupting agents brivanib, combretastatin A4 phosphate (CA4P), and 6´-sialylgalactose (SG) via semi-quantitative brightfield and qualitative confocal laser scanning microscopic (CLSM) image analysis. Compared to the reported data on in vivo efficacy of these drugs in terms of antiangiogenic and vascular disrupting effects, we observed similar trends with our 3D model, which are not reflected in conventional in vitro angiogenesis assays. High-vascular disruption under continuous treatment of the matured vascular-like network was observed at concentrations ≥3.5 ng·ml−1 for CA4P and ≥300 nM for brivanib. In contrast, SG failed to induce any significant vascular disruption in vitro. This advanced model of a 3D vascular-like network allows for testing single and combinational antiangiogenic and vascular disrupting effects with optimized dosing and may thus bridge the gap between the in vitro and in vivo experiments in validating hits from high-throughput screening. Moreover, the physiological 3D environment mimicking in vitro assay is not only highly relevant to in vivo studies linked to cancer but also to the field of tissue regeneration.
Collapse
Affiliation(s)
| | - Anna Laporte
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Inga Pacharzina
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Anke Hoppensack
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
- *Correspondence: Marie Weinhart, ,
| |
Collapse
|
41
|
Liu Z, Zhang Y, Shen N, Sun J, Tang Z, Chen X. Destruction of tumor vasculature by vascular disrupting agents in overcoming the limitation of EPR effect. Adv Drug Deliv Rev 2022; 183:114138. [PMID: 35143895 DOI: 10.1016/j.addr.2022.114138] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/27/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023]
Abstract
Nanomedicine greatly improves the efficiency in the delivery of antitumor drugs into the tumor, but insufficient tumoral penetration impairs the therapeutic efficacy of most nanomedicines. Vascular disrupting agent (VDA) nanomedicines are distributed around the tumor vessels due to the low tissue penetration in solid tumors, and the released drugs can selectively destroy immature tumor vessels and block the supply of oxygen and nutrients, leading to the internal necrosis of the tumors. VDAs can also improve the vascular permeability of the tumor, further increasing the extravasation of VDA nanomedicines in the tumor site, markedly reducing the dependence of nanomedicines on the enhanced permeability and retention effect (EPR effect). This review highlights the progress of VDA nanomedicines in recent years and their application in cancer therapy. First, the mechanisms of different VDAs are introduced. Subsequently, different strategies of delivering VDAs are described. Finally, multiple combination strategies with VDA nanomedicines in cancer therapy are described in detail.
Collapse
|
42
|
de Freitas Rego Y, Morais Costa NE, Martins de Lacerda R, Faleiros da Silva Maia A, Moreira da Silva C, de Fátima Â. Anticancer properties of arylchromenes and arylchromans: an overview. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2021-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Cancers are a set of pathologies originated by cells that have the ability to divide and multiply uncontrollably, associated with the capacity to invade and colonize adjacent tissues. Chemotherapy is one of the main approaches of treatment for cancer patients. Despite of the numerous antineoplastic drugs available, cancer cannot be cured; particularly at the late stages deprived of any side effect. Arylchromenes and arylchromans are a group of small molecules, of natural or synthetic origin, of great interest as prototypes for the drug development, especially against cancer. In this chapter, we will present the antineoplastic activity studies of the most promising examples of these arylchromenes and arylchroman derivatives.
Collapse
Affiliation(s)
- Yuri de Freitas Rego
- Departamento de Química, Grupo de Estudos em Química Orgânica e Biológica (GEQOB) , Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| | - Nathália Evelyn Morais Costa
- Departamento de Química, Grupo de Estudos em Química Orgânica e Biológica (GEQOB) , Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| | - Rodrigo Martins de Lacerda
- Departamento de Química, Grupo de Estudos em Química Orgânica e Biológica (GEQOB) , Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| | - Angélica Faleiros da Silva Maia
- Departamento de Ensino, Pesquisa e Extensão - CCO , Instituto Federal do Amazonas - Campus Coari (IFAM/CCO) , Coari , AM , Brazil
| | - Cleiton Moreira da Silva
- Departamento de Química, Grupo de Estudos em Química Orgânica e Biológica (GEQOB) , Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| | - Ângelo de Fátima
- Departamento de Química, Grupo de Estudos em Química Orgânica e Biológica (GEQOB) , Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG) , Belo Horizonte , MG , Brazil
| |
Collapse
|
43
|
Zhu D, Li Y, Zhang Z, Xue Z, Hua Z, Luo X, Zhao T, Lu C, Liu Y. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J Nanobiotechnology 2021; 19:435. [PMID: 34930293 PMCID: PMC8686559 DOI: 10.1186/s12951-021-01190-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor vessels can provide oxygen and nutrition for solid tumor tissue, create abnormal tumor microenvironment (TME), and play a vital role in the development, immune escape, metastasis and drug resistance of tumor. Tumor vessel-targeting therapy has become an important and promising direction in anti-tumor therapy, with the development of five anti-tumor therapeutic strategies, including vascular disruption, anti-angiogenesis, vascular blockade, vascular normalization and breaking immunosuppressive TME. However, the insufficient drug accumulation and severe side effects of vessel-targeting drugs limit their development in clinical application. Nanotechnology offers an excellent platform with flexible modified surface that can precisely deliver diverse cargoes, optimize efficacy, reduce side effects, and realize the combined therapy. Various nanomedicines (NMs) have been developed to target abnormal tumor vessels and specific TME to achieve more efficient vessel-targeting therapy. The article reviews tumor vascular abnormalities and the resulting abnormal microenvironment, the application of NMs in the tumor vessel-targeting strategies, and how NMs can improve these strategies and achieve multi-strategies combination to maximize anti-tumor effects. ![]()
Collapse
Affiliation(s)
- Dongjie Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
44
|
Paidakula S, Nerella S, Kankala S, Kankala RK. Recent Trends in Tubulin-Binding Combretastatin A-4 Analogs for Anticancer Drug Development. Curr Med Chem 2021; 29:3748-3773. [PMID: 34856892 DOI: 10.2174/0929867328666211202101641] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/20/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022]
Abstract
Although significant progress over several decades has been evidenced in cancer therapy, there still remains a need for the development of novel and effective therapeutic strategies to treat several relapsed and intractable cancers. In this regard, tubulin protein has become one of the efficient and major targets for anticancer drug discovery. Considering the antimitotic ability, several tubulin inhibitors have been developed to act against various cancers. Among various tubulin inhibitors available, combretastatin-A4 (CA-4), a naturally occurring lead molecule, offers exceptional cytotoxicity (including the drug-resistant cell lines) and antivascular effects. Although CA-4 offers exceptional therapeutic efficacy, several new advancements have been proposed, such as the structural modification via A and B rings, as well as cis-olefinic bridging, which provide highly efficient analogs with improved tubulin-binding efficiency to meet the anticancer drug development requirements. This review systematically emphasizes the recent trends and latest developments in the anticancer drug design & discovery, using CA-4 analogs as the tubulin inhibiting agents, highlighting their structure-activity relationships (SAR) and resultant pharmacological efficacies.
Collapse
Affiliation(s)
- Suresh Paidakula
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Srinivas Nerella
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Shravankumar Kankala
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | | |
Collapse
|
45
|
Wordeman L, Vicente JJ. Microtubule Targeting Agents in Disease: Classic Drugs, Novel Roles. Cancers (Basel) 2021; 13:5650. [PMID: 34830812 PMCID: PMC8616087 DOI: 10.3390/cancers13225650] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Microtubule-targeting agents (MTAs) represent one of the most successful first-line therapies prescribed for cancer treatment. They interfere with microtubule (MT) dynamics by either stabilizing or destabilizing MTs, and in culture, they are believed to kill cells via apoptosis after eliciting mitotic arrest, among other mechanisms. This classical view of MTA therapies persisted for many years. However, the limited success of drugs specifically targeting mitotic proteins, and the slow growing rate of most human tumors forces a reevaluation of the mechanism of action of MTAs. Studies from the last decade suggest that the killing efficiency of MTAs arises from a combination of interphase and mitotic effects. Moreover, MTs have also been implicated in other therapeutically relevant activities, such as decreasing angiogenesis, blocking cell migration, reducing metastasis, and activating innate immunity to promote proinflammatory responses. Two key problems associated with MTA therapy are acquired drug resistance and systemic toxicity. Accordingly, novel and effective MTAs are being designed with an eye toward reducing toxicity without compromising efficacy or promoting resistance. Here, we will review the mechanism of action of MTAs, the signaling pathways they affect, their impact on cancer and other illnesses, and the promising new therapeutic applications of these classic drugs.
Collapse
Affiliation(s)
| | - Juan Jesus Vicente
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA;
| |
Collapse
|
46
|
Seebacher NA, Krchniakova M, Stacy AE, Skoda J, Jansson PJ. Tumour Microenvironment Stress Promotes the Development of Drug Resistance. Antioxidants (Basel) 2021; 10:1801. [PMID: 34829672 PMCID: PMC8615091 DOI: 10.3390/antiox10111801] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Multi-drug resistance (MDR) is a leading cause of cancer-related death, and it continues to be a major barrier to cancer treatment. The tumour microenvironment (TME) has proven to play an essential role in not only cancer progression and metastasis, but also the development of resistance to chemotherapy. Despite the significant advances in the efficacy of anti-cancer therapies, the development of drug resistance remains a major impediment to therapeutic success. This review highlights the interplay between various factors within the TME that collectively initiate or propagate MDR. The key TME-mediated mechanisms of MDR regulation that will be discussed herein include (1) altered metabolic processing and the reactive oxygen species (ROS)-hypoxia inducible factor (HIF) axis; (2) changes in stromal cells; (3) increased cancer cell survival via autophagy and failure of apoptosis; (4) altered drug delivery, uptake, or efflux and (5) the induction of a cancer stem cell (CSC) phenotype. The review also discusses thought-provoking ideas that may assist in overcoming the TME-induced MDR. We conclude that stressors from the TME and exposure to chemotherapeutic agents are strongly linked to the development of MDR in cancer cells. Therefore, there remains a vast area for potential research to further elicit the interplay between factors existing both within and outside the TME. Elucidating the mechanisms within this network is essential for developing new therapeutic strategies that are less prone to failure due to the development of resistance in cancer cells.
Collapse
Affiliation(s)
| | - Maria Krchniakova
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Alexandra E. Stacy
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Patric J. Jansson
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia
| |
Collapse
|
47
|
Tsai P, Hu G, Huang P, Jheng H, Lan C, Chen Y, Chang J, Chuang S, Huang J. Design, synthesis, and in vitro/vivo anticancer activity of 4‐substituted 7‐(3‐fluoro‐4‐methoxybenzyl)‐
7
H
‐pyrrolo[2,3‐
d
]pyrimidines. J CHIN CHEM SOC-TAIP 2021. [DOI: 10.1002/jccs.202100100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Pei‐Yi Tsai
- Development Center for Biotechnology National Biotechnology Research Park Taipei City Taiwan
| | - Gong‐Siang Hu
- Department of Applied Chemistry National Chiayi University Chiayi City Taiwan
| | - Po‐Hsun Huang
- Department of Applied Chemistry National Chiayi University Chiayi City Taiwan
| | - Huei‐Lin Jheng
- Development Center for Biotechnology National Biotechnology Research Park Taipei City Taiwan
| | - Chi‐Hsuan Lan
- Department of Applied Chemistry National Chiayi University Chiayi City Taiwan
- The Training and Research Institute of Food and Agriculture National Chiayi University Chiayi City Taiwan
| | - You‐Sin Chen
- Department of Applied Chemistry National Chiayi University Chiayi City Taiwan
- The Training and Research Institute of Food and Agriculture National Chiayi University Chiayi City Taiwan
| | - Jia‐Ming Chang
- Development Center for Biotechnology National Biotechnology Research Park Taipei City Taiwan
| | - Shih‐Hsien Chuang
- Development Center for Biotechnology National Biotechnology Research Park Taipei City Taiwan
| | - Jiann‐Jyh Huang
- Department of Applied Chemistry National Chiayi University Chiayi City Taiwan
- The Training and Research Institute of Food and Agriculture National Chiayi University Chiayi City Taiwan
| |
Collapse
|
48
|
Oezdemir I, Li J, Song J, Hoyt K. 3-D Super-Resolution Ultrasound Imaging for Monitoring Early Changes in Breast Cancer after Treatment with a Vascular-Disrupting Agent. IEEE INTERNATIONAL ULTRASONICS SYMPOSIUM : [PROCEEDINGS]. IEEE INTERNATIONAL ULTRASONICS SYMPOSIUM 2021; 2021:10.1109/IUS52206.2021.9593426. [PMID: 38351971 PMCID: PMC10863700 DOI: 10.1109/ius52206.2021.9593426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The purpose of this research project was to evaluate the use of 3-dimensional (3-D) super-resolution ultrasound (SR-US) imaging to assess any early changes in breast cancer after treatment with a vascular-disrupting agent (VDA). A Vevo 3100 ultrasound system (FUJIFILM VisualSonics Inc) equipped with an MX 201 transducer was used for image acquisition. A total of 2.5 × 107 microbubbles (MBs) were injected into the tail vein of anesthetized breast cancer-bearing mice using repeat bolus injections every 5 min. A total of 10 stacks of ultrasound images were collected as the transducer was mechanically moved across the tumor at 0.6 mm intervals yielding a 6-mm thick volume. At each tumor location, a stack contained 1 × 104 frames of ultrasound data that were acquired at 463 frames/sec and stored as in-phase/quadrature (IQ) format. After motion correction, each temporal stack of ultrasound images was processed separately for clutter signal removal, which was followed by MB localization and enumeration before generation of the final SR-US image. After reconstruction of the 3-D SR-US volume dataset, the tumor microvasculature was enhanced using a multiscale vessel enhancement filter. Vessels from the resultant microvascular network were then segmented using an adaptive thresholding method. Finally, mean microvascular density (MVD) measurements from each tumor volume were computed as a summarizing statistic. While no differences were found between baseline SR-US image-derived measures of MVD (p = 0.76), these same measurements were significantly lower at 24 h after VDA treatment (p < 0.001). Overall, 3-D SR-US imaging detected early tumor changes following treatment with a vascular-targeted drug.
Collapse
Affiliation(s)
- Ipek Oezdemir
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Junjie Li
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Jane Song
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
49
|
Dai G, Zheng D, Liu G, Song Q. Synergistic Anticancer Effects of Cisplatin Combined with Combretastatin A4 Phosphate on Human Osteosarcoma-Xenografted Mice. Cells Tissues Organs 2021; 210:293-300. [PMID: 34433168 DOI: 10.1159/000517446] [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: 02/16/2021] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
This study aimed to investigate the effectiveness of anticancer therapy combining a cytotoxic chemotherapeutic agent, cisplatin (DDP), and a vascular disruptive drug, combretastatin A4 phosphate (CA4P), in osteosarcoma. First, a human osteosarcoma-xenografted mice model was established. Second, the transplanted tumor models were treated with DDP and CA4P in combination or as monotherapy. Third, the therapeutic effects and the mechanism of the drug combination in the inhibition of transplanted tumors was studied. Finally, the toxic effects of the drugs were observed and recorded. The results showed that DDP combined with CA4P significantly inhibited the growth and lung metastasis of transplanted tumors compared with the monotherapy drug group and vehicle control group. Histopathological analysis revealed that apoptotic and necrotic cell death significantly increased in the combination group, and combined treatment significantly inhibited the proliferation of osteosarcoma cells compared with either agent alone or the vehicle control. Additionally, no obvious toxic effects were observed in the combination group. These results indicate that the combined effects of DDP and CA4P on the progression of human osteosarcoma in vivo were superior to that of either agent alone. DDP combined with CA4P exerted synergistic effects at lower concentrations and promoted apoptosis and necrosis, as well as inhibited proliferation of osteosarcoma cells, but it did not increase the systemic toxic effects of chemotherapy. Our findings highlight CA4P as an effective anticancer agent candidate for combination with DDP in clinical applications to treat osteosarcoma.
Collapse
Affiliation(s)
- Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaiwei Liu
- Department of Orthopedics, Jingzhou Central Hospital, Jingzhou, China
| | - Qi Song
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, China
| |
Collapse
|
50
|
Huang L, Huang J, Nie H, Li Y, Song L, Wu F. Design, synthesis and biological evaluation of combretastatin A-4 sulfamate derivatives as potential anti-cancer agents. RSC Med Chem 2021; 12:1374-1380. [PMID: 34458740 PMCID: PMC8372205 DOI: 10.1039/d0md00372g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/17/2021] [Indexed: 01/20/2023] Open
Abstract
A series of combretastatin A-4 (CA-4) sulfamate derivatives were synthesized and their structure-activity relationship on tubulin, arylsulfatase and tumor cell antiproliferation inhibition was studied. Among them, compound 16a showed excellent potency as well as CA-4 under the same conditions against six tumor cells including HTC-116, HeLa, HepG2, MGC803, MKN45 and MCF-7 cells, respectively. Molecular docking revealed that several important hydrogen bond interactions were formed between the sulfamate group of 16a and the colchicine binding site of tubulin and steroid sulfatase respectively. Although compound 16a was less active than CA-4 in regard to its in vitro activity as an inhibitor of tubulin polymerization, it was effective as an inhibitor of arylsulfatase. This novel combretastatin A-4 sulfamate derivative has the potential to be developed as a dual inhibitor of tubulin polymerization and arylsulfatase for cancer therapy.
Collapse
Affiliation(s)
- Leilei Huang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Jinwen Huang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry CAS China
| | - Hui Nie
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Yingzi Li
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Lixing Song
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Fanhong Wu
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology China
| |
Collapse
|