1
|
Singh SB. Discovery, synthesis, activities, structure-activity relationships, and clinical development of combretastatins and analogs as anticancer drugs. A comprehensive review. Nat Prod Rep 2024; 41:298-322. [PMID: 38009216 DOI: 10.1039/d3np00053b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
Covering: 1982 to up to the end of 2022Bioassay guided purification of the extracts of Combretum caffrum led to the discovery of six series of combretastatins A-D with cytotoxic activities ranging from sub nM to >50 μM ED50's against a wide variety of cancer cell lines. Of these, cis-stilbenes combretastatins A-4 and A-1 were the most potent, exhibiting in vivo efficacy against a wide variety of tumor types in murine models. These antimitotic agents inhibited tubulin polymerization by reversibly binding to the colchicine binding sites. They inhibited tumor growth by a novel antivascular and antineogenesis mechanism in which they stopped blood flows to the blood vessels causing necrosis. Over 20 clinical trials of the phosphate prodrugs of combretastatin A-4 (CA4P) and A-1 (CA1P) showed objective and stable responses against many tumor types, with increased survival times of many patients along with the confirmed cure of certain patients inflicted with anaplastic thyroid cancers. Medicinal chemistry efforts led to the identification of three new leads (AVE8062, BNC105P, SCB01A) with improved in vitro and in vivo potency and an often-improved cellular spectrum. Unfortunately, these preclinical improvements did not translate clinically in any meaningful way. Objectively, CA4P remained the best compound and has garnered many Orphan drug designations by FDA. Clinical trials with tumor genetic mapping, particularly from previous responders, may help boost the success of these compounds in future studies. A comprehensive review of combretastatin series A-D, including bioassay guided discovery, total syntheses, and structure-activity relationship (SAR) studies, biological and mechanistic studies, and preclinical and clinical evaluations of the isolated combretastatins and analogs, along with the personal perspective of the author who originated this project, is presented.
Collapse
Affiliation(s)
- Sheo B Singh
- Charles A Dana Research Institute for Scientists Emeriti (RISE), Drew University, Madison, NJ 07940, USA.
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA.
- SBS Pharma Consulting LLC, Edison, NJ 08820, USA
| |
Collapse
|
2
|
Prasher P, Mall T, Sharma M. Synthesis and biological profile of benzoxazolone derivatives. Arch Pharm (Weinheim) 2023; 356:e2300245. [PMID: 37379239 DOI: 10.1002/ardp.202300245] [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: 05/04/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
The benzoxazolone nucleus is an ideal scaffold for drug design, owing to its discrete physicochemical profile, bioisosteric preference over pharmacokinetically weaker moieties, weakly acidic behavior, presence of both lipophilic and hydrophilic fragments on a single framework, and a wider choice of chemical modification on the benzene and oxazolone rings. These properties apparently influence the interactions of benzoxazolone-based derivatives with their respective biological targets. Hence, the benzoxazolone ring is implicated in the synthesis and development of pharmaceuticals with a diverse biological profile ranging from anticancer, analgesics, insecticides, anti-inflammatory, and neuroprotective agents. This has further led to the commercialization of several benzoxazolone-based molecules and a few others under clinical trials. Nevertheless, the SAR exploration of benzoxazolone derivatives for the identification of potential "hits" followed by the screening of "leads" provides a plethora of opportunities for further exploration of the pharmacological profile of the benzoxazolone nucleus. In this review, we aim to present the biological profile of different derivatives based on the benzoxazolone framework.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Tanisqa Mall
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, India
| |
Collapse
|
3
|
Watson EE, Russo F, Moreau D, Winssinger N. Optochemical Control of Therapeutic Agents through Photocatalyzed Isomerization. Angew Chem Int Ed Engl 2022; 61:e202203390. [PMID: 35510306 PMCID: PMC9400970 DOI: 10.1002/anie.202203390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 12/04/2022]
Abstract
A Ru(bpy)3Cl2 photocatalyst is applied to the rapid trans to cis isomerization of a range of alkene‐containing pharmacological agents, including combretastatin A‐4 (CA‐4), a clinical candidate in oncology, and resveratrol derivatives, switching their configuration from inactive substances to potent cytotoxic agents. Selective in cellulo activation of the CA‐4 analog Res‐3M is demonstrated, along with its potent cytotoxicity and inhibition of microtubule dynamics.
Collapse
Affiliation(s)
- Emma E. Watson
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| | - Francesco Russo
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| | - Dimitri Moreau
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| | - Nicolas Winssinger
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| |
Collapse
|
4
|
Watson EE, Russo F, Moreau D, Winssinger N. Optochemical Control of Therapeutic Agents through Photocatalyzed Isomerization. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202203390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Emma E. Watson
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| | - Francesco Russo
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| | - Dimitri Moreau
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| | - Nicolas Winssinger
- Department of Organic Chemistry NCCR Chemical Biology Faculty of Sciences University of Geneva 1211 Geneva Switzerland
| |
Collapse
|
5
|
The Proper Administration Sequence of Radiotherapy and Anti-Vascular Agent-DMXAA Is Essential to Inhibit the Growth of Melanoma Tumors. Cancers (Basel) 2021; 13:cancers13163924. [PMID: 34439079 PMCID: PMC8394873 DOI: 10.3390/cancers13163924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023] Open
Abstract
Vascular disrupting agents (VDAs), such as DMXAA, effectively destroy tumor blood vessels and cause the formation of large areas of necrosis in the central parts of the tumors. However, the use of VDAs is associated with hypoxia activation and residues of rim cells on the edge of the tumor that are responsible for tumor regrowth. The aim of the study was to combine DMXAA with radiotherapy (brachytherapy) and find the appropriate administration sequence to obtain the maximum synergistic therapeutic effect. We show that the combination in which tumors were irradiated prior to VDAs administration is more effective in murine melanoma growth inhibition than in either of the agents individually or in reverse combination. For the first time, the significance of immune cells' activation in such a combination is demonstrated. The inhibition of tumor growth is linked to the reduction of tumor blood vessels, the increased infiltration of CD8+ cytotoxic T lymphocytes and NK cells and the polarization of macrophages to the cytotoxic M1 phenotype. The reverse combination of therapeutic agents showed no therapeutic effect and even abolished the effect of DMXAA. The combination of brachytherapy and vascular disrupting agent effectively inhibits the growth of melanoma tumors but requires careful planning of the sequence of administration of the agents.
Collapse
|
6
|
Smolarczyk R, Czapla J, Jarosz-Biej M, Czerwinski K, Cichoń T. Vascular disrupting agents in cancer therapy. Eur J Pharmacol 2020; 891:173692. [PMID: 33130277 DOI: 10.1016/j.ejphar.2020.173692] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Tumor blood vessel formation is a key process for tumor expansion. Tumor vessels are abnormal and differ from normal vessels in architecture and components. Besides oxygen and nutrients supply, the tumor vessels system, due to its abnormality, is responsible for: hypoxia formation, and metastatic routes. Tumor blood vessels can be a target of anti-cancer therapies. There are two types of therapies that target tumor vessels. The first one is the inhibition of the angiogenesis process. However, the inhibition is often ineffective because of alternative angiogenesis mechanism activation. The second type is a specific targeting of existing tumor blood vessels by vascular disruptive agents (VDAs). There are three groups of VDAs: microtubule destabilizing drugs, flavonoids with anti-vascular functions, and tumor vascular targeted drugs based on endothelial cell receptors. However, VDAs possess some limitations. They may be cardiotoxic and their application in therapy may leave viable residual, so called, rim cells on the edge of the tumor. However, it seems that a well-designed combination of VDAs with other anti-cancer drugs may bring a significant therapeutic effect. In this article, we describe three groups of vascular disruptive agents with their advantages and disadvantages. We mention VDAs clinical trials. Finally, we present the current possibilities of VDAs combination with other anti-cancer drugs.
Collapse
Affiliation(s)
- 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.
| | - Justyna Czapla
- 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.
| | - Magdalena Jarosz-Biej
- 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.
| | - Kyle Czerwinski
- University of Manitoba, Faculty of Science. 66 Chancellors Cir, Winnipeg, Canada.
| | - Tomasz Cichoń
- 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
|
7
|
Tewari KS, Sill MW, Coleman RL, Aghajanian C, Mannel R, DiSilvestro PA, Powell M, Randall LM, Farley J, Rubin SC, Monk BJ. Bevacizumab plus fosbretabulin in recurrent ovarian cancer: Overall survival and exploratory analyses of a randomized phase II NRG oncology/gynecologic oncology group study. Gynecol Oncol 2020; 159:79-87. [PMID: 32723679 DOI: 10.1016/j.ygyno.2020.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/07/2020] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To explore the relationship between tumor size and response to combined anti-vascular targeted therapy using the anti-angiogenesis inhibitor, bevacizumab, and the tubulin-binding vascular disrupting agent, fosbretabulin. METHODS An exploratory, post-hoc analysis of the randomized phase II trial, Gynecologic Oncology Group-0186I, was performed. One hundred and seven patients with recurrent ovarian carcinoma, treated with up to 3 prior regimens, were randomized to bevacizumab 15 mg/kg body weight with or without intravenous fosbretabulin 60 mg/m2 body surface area every 21 days until progression or unacceptable toxicity. The primary analysis favored the combination (HR 0.69; 95% CI, 0.47-1.00; p = .049) [Monk BJ, et al. J Clin Oncol 2016;34:2279-86]. The Cox proportional hazards model was used to estimate the treatment effect in various subpopulations. RESULTS With extended follow-up, the median PFS for bevacizumab plus fosbretabulin was 7.6 months as compared to 4.8 months with bevacizumab alone (HR 0.74; 90% CI, 0.54-1.02). Overall survival was similar in the experimental and control arms (25.2 vs 24.4 mos, respectively, HR 0.85; 90% CI, 0.59-1.22; p = .461). Eighty-one patients had measurable disease and median tumor size was 5.7 cm. In the ≤5.7 cm subgroup, the HR for progression or death was 0.77 (90% CI 0.45-1.31). Patients with tumors >5.7 cm (n = 40) had a HR for progression or death of 0.55; 90% CI, 0.32-0.96; p = .075). CONCLUSIONS Although no significant survival benefit was observed, the trend showing a reduced HR for progression or death with increasing tumor size when fosbretabulin is added to bevacizumab compared to bevacizumab alone warrants further study.
Collapse
Affiliation(s)
- Krishnansu S Tewari
- University of California, Irvine, Division of Gynecologic Oncology, Orange, CA, United States.
| | - Michael W Sill
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park, Buffalo, NY 14263, United States.
| | | | - Carol Aghajanian
- Memorial Sloan-Kettering Cancer Center, New York, NY, United States.
| | - Robert Mannel
- The Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, 800 NE 10(th) St., SOCC 6043, Oklahoma City, OK 73104, United States.
| | - Paul A DiSilvestro
- Women and Infants Hospital of Rhode Island, 101 Dudley St., Providence, RI 02905, United States.
| | - Matthew Powell
- Washington University School of Medicine, 4911 Barnes Jewish Hospital Plaza, St. Louis, MO 63110, United States.
| | - Leslie M Randall
- University of California, Irvine, Division of Gynecologic Oncology, Orange, CA, United States.
| | - John Farley
- Creighton University School of Medicine at St. Joseph's Hospital and Medical Center, Pheonix, AZ 85013, United States.
| | - Stephen C Rubin
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111, United States.
| | - Bradley J Monk
- Arizona Oncology (US Oncology Network), University of Arizona College of Medicine-Phoenix, Creighton University School of Medicine, St. Joseph's Hospital, Phoenix, AZ, United States.
| |
Collapse
|
8
|
Predicting Clinical Efficacy of Vascular Disrupting Agents in Rodent Models of Primary and Secondary Liver Cancers: An Overview with Imaging-Histopathology Correlation. Diagnostics (Basel) 2020; 10:diagnostics10020078. [PMID: 32024029 PMCID: PMC7168934 DOI: 10.3390/diagnostics10020078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
Vascular disrupting agents (VDAs) have entered clinical trials for over 15 years. As the leading VDA, combretastatin A4 phosphate (CA4P) has been evaluated in combination with chemotherapy and molecular targeting agents among patients with ovarian cancer, lung cancer and thyroid cancer, but still remains rarely explored in human liver cancers. To overcome tumor residues and regrowth after CA4P monotherapy, a novel dual targeting pan-anticancer theragnostic strategy, i.e., OncoCiDia, has been developed and shown promise previously in secondary liver tumor models. Animal model of primary liver cancer is time consuming to induce, but of value for more closely mimicking human liver cancers in terms of tumor angiogenesis, histopathological heterogeneity, cellular differentiation, tumor components, cancer progression and therapeutic response. Being increasingly adopted in VDA researches, multiparametric magnetic resonance imaging (MRI) provides imaging biomarkers to reflect in vivo tumor responses to drugs. In this article as a chapter of a doctoral thesis, we overview the construction and clinical relevance of primary and secondary liver cancer models in rodents. Target selection for CA4P therapy assisted by enhanced MRI using hepatobiliary contrast agents (CAs), and therapeutic efficacy evaluated by using MRI with a non-specific contrast agent, dynamic contrast enhanced (DCE) imaging, diffusion weighted imaging (DWI) are also described. We then summarize diverse responses among primary hepatocellular carcinomas (HCCs), secondary liver and pancreatic tumors to CA4P, which appeared to be related to tumor size, vascularity, and cellular differentiation. In general, imaging-histopathology correlation studies allow to conclude that CA4P tends to be more effective in secondary liver tumors and in more differentiated HCCs, but less effective in less differentiated HCCs and implanted pancreatic tumor. Notably, cirrhotic liver may be responsive to CA4P as well. All these could be instructive for future clinical trials of VDAs.
Collapse
|
9
|
Abma E, De Spiegelaere W, Vanderperren K, Stock E, Van Brantegem L, Cornelis I, Daminet S, Ni Y, Vynck M, Verstraete G, Smets P, de Rooster H. A single dose of intravenous combretastatin A4-phosphate is reasonably well tolerated and significantly reduces tumour vascularization in canine spontaneous cancers. Vet Comp Oncol 2018; 16:467-477. [PMID: 29797763 DOI: 10.1111/vco.12402] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 12/01/2022]
Abstract
Combretastatin A4-phosphate (CA4P) is an anti-tumour vascular targeting agent which selectively blocks tumour blood flow. Research on CA4P in rodent tumour models is extensive; however, knowledge of its effect on spontaneous cancer is scarce. This study was conducted in canine patients with spontaneous solid tumours. The goal was to assess the toxicity and efficacy of CA4P in various spontaneous tumour types. Eight dogs with spontaneous tumours were enrolled and treated with a single dose of 75 mg m-2 intravenous CA4P. The dogs were screened and monitored before and after injection. Pre- and post-treatment tumour blood flow was analysed in vivo by power Doppler ultrasound (PDUS) and contrast-enhanced ultrasound (CEUS). Vessel destruction and tumour necrosis were evaluated by histopathology. Clinically relevant toxicity was limited to one case of temporary tetraparesis; other adverse events were mild. Significant cardiovascular changes were mostly confined to changes in heart rate and cTnI levels. Macroscopic tumour size reduction was evident in 2 dogs. Based on PDUS and CEUS, CA4P induced a significant decrease in vascular index and tumour blood flow. Post-treatment, histopathology revealed a significant increase of necrotic tumoural tissue and a significant reduction in microvessel density in tumoural tissue. Anti-vascular and necrotizing effects of CA4P were documented in a variety of canine spontaneous cancers with only minimal side effects. This is the first study reporting the administration of CA4P to canine cancer patients with in vivo and ex vivo assessment, and a first step toward implementing CA4P in combination therapies in veterinary oncology patients. The use of CA4P in canine patients was approved and registered by the Belgian Federal Agency for Medicines and Health Products (FAMHP) (approval number 0002588, registration number 6518 ID 2F12).
Collapse
Affiliation(s)
- E Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital Ghent, Ghent, Belgium
| | - W De Spiegelaere
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - K Vanderperren
- Department of Medical Imaging and Orthopedics of Small Animals, Faculty of Veterinary Medicine, University of Ghent, Ghent, Belgium
| | - E Stock
- Department of Medical Imaging and Orthopedics of Small Animals, Faculty of Veterinary Medicine, University of Ghent, Ghent, Belgium
| | - L Van Brantegem
- Department of Pathology, Bacteriology and Poultry Diseases, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - I Cornelis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - S Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Y Ni
- Theragnostic Lab, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - M Vynck
- Department of Data Analysis and Mathematical Modeling, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - G Verstraete
- Laboratory of Pharmaceutical Technology, Ghent University, Ghent, Belgium
| | - P Smets
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - H de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital Ghent, Ghent, Belgium
| |
Collapse
|
10
|
Siemann DW, Chaplin DJ, Horsman MR. Realizing the Potential of Vascular Targeted Therapy: The Rationale for Combining Vascular Disrupting Agents and Anti-Angiogenic Agents to Treat Cancer. Cancer Invest 2017; 35:519-534. [DOI: 10.1080/07357907.2017.1364745] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- D. W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | | - M. R. Horsman
- Department of Experimental Clinical Oncology, Aarhus University, Denmark
| |
Collapse
|
11
|
Liu Y, Yin T, Keyzer FD, Feng Y, Chen F, Liu J, Song S, Yu J, Vandecaveye V, Swinnen J, Bormans G, Himmelreich U, Oyen R, Zhang J, Huang G, Ni Y. Micro-HCCs in rats with liver cirrhosis: paradoxical targeting effects with vascular disrupting agent CA4P. Oncotarget 2017; 8:55204-55215. [PMID: 28903414 PMCID: PMC5589653 DOI: 10.18632/oncotarget.19339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
We sought to investigate anticancer efficacy of a vascular disrupting agent (VDA) combretastatin A-4 phosphate (CA4P) in relation to tumor size among hepatocellular carcinomas (HCCs) in rats using magnetic resonance imaging (MRI) and postmortem techniques. Nineteen rats with 43 chemically-induced HCCs of 2.8–20.9 mm in size on liver cirrhosis received CA4P intravenously at 10 mg/kg. Tumor-diameter was measured by T2-weighted imaging (T2WI) to define microcancers (< 5 mm) versus larger HCCs. Vascular responses and tissue necrosis were detected by diffusion-weighted imaging (DWI), contrast-enhanced T1-weighted imaging (CE-T1WI) and dynamic contrast enhanced (DCE-) MRI, which were validated by microangiography and histopathology. MRI revealed nearly complete necrosis in 5 out of 7 micro-HCCs, but diverse therapeutic necrosis in larger HCCs with a positive correlation with tumor size. Necrosis in micro-HCCs was 36.9% more than that in larger HCCs. While increased diffusion coefficient (ADCdiff) suggested tumor necrosis, perfusion coefficient (ADCperf) indicated sharply decreased blood perfusion in cirrhotic liver together with a reduction in micro-HCCs. DCE revealed lowered tumor blood flow from intravascular into extravascular extracellular space (EES). Microangiography and histopathology revealed hypo- and hypervascularity in 4 and 3 micro-HCCs, massive, partial and minor degrees of tumoral necrosis in 5, 1 and 1 micro-HCCs respectively, and patchy necrotic foci in cirrhotic liver. CD34-PAS staining implicated that poorly vascularized micro-HCCs growing on liver cirrhosis tended to respond better to CA4P treatment. In this study, more complete CA4P-response occurred unexpectedly in micro-HCCs in rats, along with CA4P-induced necrotic foci in cirrhotic liver. These may help to plan clinical applications of VDAs in patients with HCCs and liver cirrhosis.
Collapse
Affiliation(s)
- Yewei Liu
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium.,Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.,Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | - Ting Yin
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | | | - Yuanbo Feng
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Feng Chen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jianjun Liu
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shaoli Song
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jie Yu
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | | | - Johan Swinnen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Guy Bormans
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Uwe Himmelreich
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Raymond Oyen
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jian Zhang
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Gang Huang
- Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.,Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | - Yicheng Ni
- Biomedical Group, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
12
|
Chase DM, Chaplin DJ, Monk BJ. The development and use of vascular targeted therapy in ovarian cancer. Gynecol Oncol 2017; 145:393-406. [DOI: 10.1016/j.ygyno.2017.01.031] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 02/08/2023]
|
13
|
Garon EB, Neidhart JD, Gabrail NY, de Oliveira MR, Balkissoon J, Kabbinavar F. A randomized Phase II trial of the tumor vascular disrupting agent CA4P (fosbretabulin tromethamine) with carboplatin, paclitaxel, and bevacizumab in advanced nonsquamous non-small-cell lung cancer. Onco Targets Ther 2016; 9:7275-7283. [PMID: 27942221 PMCID: PMC5138047 DOI: 10.2147/ott.s109186] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Combretastatin A4-phosphate, fosbretabulin tromethamine (CA4P) is a vascular disrupting agent that targets tumor vasculature. This study evaluated the safety of CA4P when combined with carboplatin, paclitaxel, and bevacizumab in chemotherapy-naïve subjects with advanced nonsquamous, non-small-cell lung cancer. Methods Adult subjects with confirmed American Joint Committee on Cancer six stage IIIB/IV non-small-cell lung cancer and an Eastern Cooperative Oncology Group performance score of 0 or 1 were randomized to receive six cycles (treatment phase) of paclitaxel (200 mg/m2), carboplatin (area under the concentration versus time curve 6), and bevacizumab (15 mg/kg) on day 1 and repeated every 21 days, or this regimen plus CA4P (60 mg/m2) on days 7, 14, and 21 of each cycle. Subjects could then receive additional maintenance treatment (excluding carboplatin and paclitaxel) for up to 1 year. Results Sixty-three subjects were randomized, 31 to control and 32 to CA4P, and 19 (61.3%) and 17 (53.1%), respectively, completed the treatment phase. Exposure to study treatment and dose modifications were comparable between the randomized groups. The overall incidence of treatment-emergent adverse events was similar between groups, with increased neutropenia, leukopenia, and hypertension in the CA4P group. Deaths, serious adverse events, and early discontinuations from treatment were comparable between the randomized treatment groups. The overall tumor response rate with CA4P was 50% versus 32% in controls. Overall and progression-free survival rates were comparable between the groups. Conclusion CA4P plus carboplatin, paclitaxel, and bevacizumab appears to be a tolerable regimen with an acceptable toxicity profile in subjects with advanced non-small-cell lung cancer.
Collapse
Affiliation(s)
- Edward B Garon
- Department of Medicine; Department of Hematology and Oncology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | | | | | | | - Jai Balkissoon
- Global Product Development and Immuno-Oncology, PPD, Wilmington, NC, USA
| | - Fairooz Kabbinavar
- Department of Medicine; Department of Hematology and Oncology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
14
|
Johnson SP, Ramasawmy R, Campbell-Washburn AE, Wells JA, Robson M, Rajkumar V, Lythgoe MF, Pedley RB, Walker-Samuel S. Acute changes in liver tumour perfusion measured non-invasively with arterial spin labelling. Br J Cancer 2016; 114:897-904. [PMID: 27031853 PMCID: PMC4984798 DOI: 10.1038/bjc.2016.51] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/26/2016] [Accepted: 02/04/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Non-invasive measures of tumour vascular perfusion are desirable, in order to assess response to vascular targeting (or modifying) therapies. In this study, hepatic arterial spin labelling (ASL) magnetic resonance imaging (MRI) was investigated to measure acute changes in perfusion of colorectal cancer in the liver, in response to vascular disruption therapy with OXi4503. METHODS SW1222 and LS174T tumours were established in the liver of MF1 nu/nu mice via intrasplenic injection. Perfusion and R2(*) MRI measurements were acquired with an Agilent 9.4T horizontal bore scanner, before and at 90 min after 40 mg kg(-1) OXi4503. RESULTS A significant decrease in SW1222 tumour perfusion was observed (-43±33%, P<0.005). LS174T tumours had a significantly lower baseline level of perfusion. Intrinsic susceptibility MRI showed a significant increase in R2(*) in LS174T tumours (28±25%, P<0.05). An association was found between the change in tumour perfusion and the proximity to large vessels, with pre-treatment blood flow predictive of subsequent response. Histological evaluation confirmed the onset of necrosis and evidence of heterogeneous response between tumour deposits. CONCLUSIONS Hepatic ASL-MRI can detect acute response to targeted tumour vascular disruption entirely non-invasively. Hepatic ASL of liver tumours has potential for use in a clinical setting.
Collapse
Affiliation(s)
- S Peter Johnson
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
- UCL Centre for Advanced Biomedical Imaging, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Rajiv Ramasawmy
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
- UCL Centre for Advanced Biomedical Imaging, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Adrienne E Campbell-Washburn
- UCL Centre for Advanced Biomedical Imaging, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
- Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jack A Wells
- UCL Centre for Advanced Biomedical Imaging, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Mathew Robson
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Vineeth Rajkumar
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - R Barbara Pedley
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Simon Walker-Samuel
- UCL Centre for Advanced Biomedical Imaging, University College London, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
15
|
Stabilizing versus destabilizing the microtubules: a double-edge sword for an effective cancer treatment option? Anal Cell Pathol (Amst) 2015; 2015:690916. [PMID: 26484003 PMCID: PMC4592889 DOI: 10.1155/2015/690916] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/29/2015] [Accepted: 09/01/2015] [Indexed: 11/17/2022] Open
Abstract
Microtubules are dynamic and structural cellular components involved in several cell functions, including cell shape, motility, and intracellular trafficking. In proliferating cells, they are essential components in the division process through the formation of the mitotic spindle. As a result of these functions, tubulin and microtubules are targets for anticancer agents. Microtubule-targeting agents can be divided into two groups: microtubule-stabilizing, and microtubule-destabilizing agents. The former bind to the tubulin polymer and stabilize microtubules, while the latter bind to the tubulin dimers and destabilize microtubules. Alteration of tubulin-microtubule equilibrium determines the disruption of the mitotic spindle, halting the cell cycle at the metaphase-anaphase transition and, eventually, resulting in cell death. Clinical application of earlier microtubule inhibitors, however, unfortunately showed several limits, such as neurological and bone marrow toxicity and the emergence of drug-resistant tumor cells. Here we review several natural and synthetic microtubule-targeting agents, which showed antitumor activity and increased efficacy in comparison to traditional drugs in various preclinical and clinical studies. Cryptophycins, combretastatins, ombrabulin, soblidotin, D-24851, epothilones and discodermolide were used in clinical trials. Some of them showed antiangiogenic and antivascular activity and others showed the ability to overcome multidrug resistance, supporting their possible use in chemotherapy.
Collapse
|
16
|
Liu L, Mason RP, Gimi B. Dynamic bioluminescence and fluorescence imaging of the effects of the antivascular agent Combretastatin-A4P (CA4P) on brain tumor xenografts. Cancer Lett 2014; 356:462-9. [PMID: 25305449 DOI: 10.1016/j.canlet.2014.09.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 02/08/2023]
Abstract
Combretastatin A-4 (CA4) is a natural product isolated from Combretum caffrum that inhibits tubulin polymerization by binding to the colchicine-binding site. A corresponding water soluble pro-drug (referred to as CA4P), has undergone extensive clinical trials and has been evaluated in pre-clinical studies using multiple modalities. We previously reported a novel assay based on dynamic bioluminescent imaging to assess tumor vascular disruption and now present its application to assessing multiple tumors simultaneously. The current study evaluated the vascular-disrupting activity of CA4P on subcutaneous 9L rat brain tumor xenografts in mice using dynamic bioluminescence imaging. A single dose of CA4P (120 mg/kg, intraperitoneally) induced rapid, temporary tumor vascular shutdown revealed by a rapid and reproducible decrease of light emission from luciferase-expressing 9L tumors following administration of luciferin as a substrate. A time-dependent reduction of tumor perfusion after CA4P treatment was confirmed by immunohistological assessment of the perfusion marker Hoechst 33342 and the tumor vasculature marker CD31. The vasculature showed distinct recovery within 24 h post therapy. Multiple tumors behaved similarly, although a size dependent vascular inhibition was observed. In conclusion, CA4P caused rapid, temporary tumor vascular shutdown and led to reduction of tumor perfusion in rat brain tumor xenografts and the multiple tumor approach should lead to more efficient studies requiring fewer animals and greater consistency.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Barjor Gimi
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
17
|
Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C, Marur S, Gramza A, Yosef RB, Gitlitz B, Haugen BR, Ondrey F, Lu C, Karandikar SM, Khuri F, Licitra L, Remick SC. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid 2014; 24:232-40. [PMID: 23721245 DOI: 10.1089/thy.2013.0078] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC), a rare highly vascularized tumor, has a dismal outcome. We conducted an open-label study of doublet carboplatin/paclitaxel chemotherapy with or without fosbretabulin in patients with ATC. METHODS Patients were randomly assigned in a 2:1 ratio to 6 cycles of paclitaxel 200 mg/m(2) followed by carboplatin AUC 6 on day 1 every 3 weeks (CP), or these drugs were given on day 2 after fosbretabulin 60 mg/m(2) (CP/fosbretabulin) on days 1, 8 and 15. After 6 cycles, patients on the fosbretabulin arm without progression could continue to receive fosbretabulin on days 1 and 8 of a 3-week schedule until progression. The primary end point was overall survival (OS). RESULTS Eighty patients were assigned (planned, 180) when enrollment was stopped due to rarity of disease and very low accrual. Median OS was 5.2 months [95% confidence interval (CI) 3.1, 9.0] for the CP/fosbretabulin arm (n=55; hazard ratio 0.73 [95% CI 0.44, 1.21]) and 4.0 months [95% CI 2.8, 6.2] for the CP arm (n=25; p=0.22 [log rank test]). One-year survival for CP/fosbretabulin versus CP was 26% versus 9%, respectively. There was no significant difference in progression-free survival between the two arms. Grade 1-2 hypertension and grade 3-4 neutropenia were more common with CP/fosbretabulin. There were no significant adverse cardiovascular side effects. CONCLUSIONS Although the study did not meet statistical significance in improvement in OS with the addition of fosbretabulin to carboplatin/paclitaxel, it represents the largest prospective randomized trial ever conducted in ATC. The regimen is well tolerated, with AEs and deaths primarily related to ATC and disease progression.
Collapse
Affiliation(s)
- Julie A Sosa
- 1 Department of Surgery, Duke University School of Medicine , Durham, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Vascular disrupting agents (VDAs) are a relatively new class of drugs that target tumor vasculature and induce tumor blood flow shutdown and subsequent necrosis in the tumor core. The first generation of these agents is actively evaluated in clinical trials, whereas new molecules are developed in order to enhance efficacy and to overcome resistance mechanisms. VDA used as a single agent only cause a moderate tumor growth delay. So, strategy aiming at combining VDA to conventional cancer treatments is undergoing extensive investigations. A special emphasis has been put on combination with chemotherapeutic agents. Besides, numerous preclinical studies have also clearly established that the association of VDA to radiotherapy can improve antitumor treatment and may lead to a therapeutic gain. However, up to date, there is a lack of clinical trials evaluating such combinations, whereas it would be of great interest since radiotherapy is widely used as anticancer treatment.
Collapse
|
19
|
Nielsen T, Murata R, Maxwell RJ, Stødkilde-Jørgensen H, Ostergaard L, Ley CD, Kristjansen PEG, Horsman MR. Non-invasive imaging of combretastatin activity in two tumor models: Association with invasive estimates. Acta Oncol 2010; 49:906-13. [PMID: 20831477 DOI: 10.3109/0284186x.2010.499135] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The efficacy of the vascular disrupting agent combretastatin A-4 phosphate (CA4P) depends on several factors including tumor size, nitric oxide level, interstitial fluid pressure, and vascular permeability. These factors vary among tumor types. The aim of this study was to investigate all these factors in two tumor models that respond differently to CA4P. MATERIAL AND METHODS Mice bearing C3H mammary carcinomas or KHT sarcomas (200 to 800 mm(3)) were intraperitoneally injected with CA4P (100 mg/kg). Tumor size and the effect of a nitric oxide inhibitor nitro-L-arginine (NLA) administered intravenously were evaluated by necrotic fraction histologically assessed at 24 hours. Interstitial fluid pressure (IFP) was measured using the wick-in-needle technique, and vascular characteristics were assessed with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). RESULTS Initial necrotic fraction was about 10% in both tumor models at 200 mm(3), but only increased significantly with tumor size in the C3H mammary carcinoma. In this tumor, CA4P significantly induced further necrosis by about 15% at all sizes, but in the KHT tumor, the induced necrotic fraction depended on tumor size. For both tumor types, NLA with CA4P significantly increased necrotic fraction above that for each drug alone. CA4P significantly decreased IFP in all tumors except in the 800 mm(3) C3H tumor, which had an initially non-significant lower value. Interstitial volume estimated by DCE-MRI increased in all groups, except the 800 mm(3) C3H tumors. DCE-MRI vascular parameters showed different initial characteristics and general significant reductions following CA4P treatment. CONCLUSIONS Both tumor models showed differences in all factors before treatment, and in their response to CA4P. Perfusion and permeability as estimated by DCE-MRI play a central role in the CA4P response, and interstitial volume and IFP seemed related. These factors may be of clinical value in the planning of CA4P treatments.
Collapse
Affiliation(s)
- Thomas Nielsen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Higa GM. The microtubule as a breast cancer target. Breast Cancer 2010; 18:103-19. [PMID: 20862571 DOI: 10.1007/s12282-010-0224-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 08/24/2010] [Indexed: 01/19/2023]
Abstract
Manifestations of non-equilibrium polarity, random transgressions, and catastrophes are not conditions usually associated with a sense of normalcy. Yet these disquieting features distinguish a utilitarian behavior known as dynamic instability, the signature characteristic of the microtubule. Long known to be a tumor target, disruption of this fragile attribute is associated with some of the most effective agents used to treat breast cancer today. Although the biology of the microtubule is under intense investigation much still remains unknown. As such, our understanding of regulatory molecules and resistance mechanisms are still rudimentary, further compromising our ability to develop novel therapeutic strategies to improve microtubule inhibitors. This review focuses on several classes of anti-microtubule agents and their effects on the functional dynamics of the targeted polymer. The primary objective is to critically examine the molecular mechanisms that contribute to tumor cell death, tumor-resistance, and incident neurotoxicity.
Collapse
Affiliation(s)
- Gerald M Higa
- Mary Babb Randolph Cancer Center, West Virginia University Schools of Pharmacy and Medicine, Morgantown, WV, USA.
| |
Collapse
|
21
|
Siemann DW. The unique characteristics of tumor vasculature and preclinical evidence for its selective disruption by Tumor-Vascular Disrupting Agents. Cancer Treat Rev 2010; 37:63-74. [PMID: 20570444 DOI: 10.1016/j.ctrv.2010.05.001] [Citation(s) in RCA: 431] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 04/28/2010] [Accepted: 05/02/2010] [Indexed: 02/06/2023]
Abstract
The vasculature of solid tumors is fundamentally different from that of normal vasculature and offers a unique target for anti-cancer therapy. Direct vascular-targeting with Tumor-Vascular Disrupting Agents (Tumor-VDAs) is distinctly different from anti-angiogenic strategies, and offers a complementary approach to standard therapies. Tumor-VDAs therefore have significant potential when combined with chemotherapy, radiotherapy, and angiogenesis-inhibiting agents. Preclinical studies with the different Tumor-VDA classes have demonstrated key tumor-selective anti-vascular and anti-tumor effects.
Collapse
Affiliation(s)
- Dietmar W Siemann
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
22
|
|
23
|
A Phase Ib trial of CA4P (combretastatin A-4 phosphate), carboplatin, and paclitaxel in patients with advanced cancer. Br J Cancer 2010; 102:1355-60. [PMID: 20389300 PMCID: PMC2865759 DOI: 10.1038/sj.bjc.6605650] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The vascular disrupting agent combretastatin A4 phosphate (CA4P) causes major regression of animal tumours when given as combination therapy. METHODS Patients with advanced cancer refractory to standard therapy were treated with CA4P as a 10-min infusion, 20 h before carboplatin, paclitaxel, or paclitaxel, followed by carboplatin. RESULTS Combretastatin A4 phosphate was escalated from 36 to 54 mg m(-2) with the carboplatin area under the concentration curve (AUC) 4-5, from 27 to 54 mg m(-2) with paclitaxel 135-175 mg m(-2), and from 54 to 72 mg m(-2) with carboplatin AUC 5 and paclitaxel 175 mg m(-2). Grade 3 or 4 neutropenia was seen in 17%, and thrombocytopenia only in 4% of 46 patients. Grade 1-3 hypertension (26% of patients) and grade 1-3 tumour pain (65% of patients) were the most typical non-haematological toxicities. Dose-limiting toxicity of grade 3 hypertension or grade 3 ataxia was seen in two patients at 72 mg m(-2). Responses were seen in 10 of 46 (22%) patients with ovarian, oesophageal, small-cell lung cancer, and melanoma. CONCLUSION The combination of CA4P with carboplatin and paclitaxel was well tolerated in the majority of patients with adequate premedication and had antitumour activity in patients who were heavily pretreated.
Collapse
|
24
|
Treatment of rodent liver tumor with combretastatin a4 phosphate: noninvasive therapeutic evaluation using multiparametric magnetic resonance imaging in correlation with microangiography and histology. Invest Radiol 2009; 44:44-53. [PMID: 19034028 DOI: 10.1097/rli.0b013e31818e5ace] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To document tumoricidal events after intravenous administration of a vascular targeting agent combretastatin A-4-phosphate (CA4P) in rodent liver tumors by using multiparametric magnetic resonance imaging (MRI) in correlation with microangiography and histopathology. MATERIALS AND METHODS Thirty rhabdomyosarcomas of 8 to 14 mm in diameter were obtained 16 days after implantation in liver lobes of 15 rats. Using a 1.5T magnet and a 4-channel wrist coil, T2-weighted imaging (T2WI), pre- and postcontrast T1-weighted imaging (T1WI), diffusion-weighted imaging (DWI), and dynamic susceptibility imaging (DSI) with relative blood volume (rBV) and flow (rBF) maps were acquired at baseline, 1 hour, 6 hours, and 48 hours after iv injection of CA4P at 10 mg/kg and vehicle in 9 treated and 6 control rats, respectively. In vivo data including signal intensity (SI), tumor volume, apparent diffusion coefficient (ADC), rBV, and rBF were correlated with ex vivo microangiographic and histopathologic findings. RESULTS CA4P-treated tumors (n = 18) grew slower than those (n = 12) of controls (P < 0.05), with vascular shutdown evident on CE-T1WI at 1 hour but more prominent at 6 hours. However, enhanced rim occurred in the periphery 48 hours after treatment, indicating neovascularization. ADC map enabled distinction between necrotic and viable tumors. DSI-derived tumoral rBV and rBF decreased significantly at 1 hour through 6 hours and partly recovered at 48 hours. SI-time curve reflected diverse therapeutic responses between tumor and liver. MRI findings were verified by ex vivo techniques. CONCLUSIONS Clinical MRI allowed monitoring of CA4P-related vascular shutdown, necrosis, and neovascularization of liver tumors in rats. Single dose of CA4P seemed insufficient for tumor eradication because of evident peripheral residue and recurrence.
Collapse
|
25
|
Ren X, Dai M, Lin LP, Li PK, Ding J. Anti-angiogenic and vascular disrupting effects of C9, a new microtubule-depolymerizing agent. Br J Pharmacol 2009; 156:1228-38. [PMID: 19302593 DOI: 10.1111/j.1476-5381.2009.00112.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE The critical role of blood supply in the growth of solid tumours makes blood vessels an ideal target for anti-tumour drug discovery. The anti-angiogenic and vascular disrupting activities of C9, a newly synthesized microtubule-depolymerizing agent, were investigated with several in vitro and in vivo models. Possible mechanisms involved in its activity were also assessed. EXPERIMENTAL APPROACH Microtubule-depolymerizing actions were assessed by surface plasmon resonance binding, competitive inhibition and cytoskeleton immunofluorescence. Anti-angiogenic and vascular disrupting activities were tested on proliferation, migration, tube formation with human umbilical vein endothelial cells, and in rat aortic ring, chick chorioallantoic membrane and Matrigel plug assays. Western blots and Rho activation assays were employed to examine the role of Raf-MEK-ERK (mitogen-activated ERK kinase, extracellular signal-regulated kinase) and Rho/Rho kinase signalling. KEY RESULTS C9 inhibited proliferation, migration and tube formation of endothelial cells and inhibited angiogenesis in aortic ring and chick chorioallantoic membrane assays. C9 induced disassembly of microtubules in endothelial cells and down-regulated Raf-MEK-ERK signalling activated by pro-angiogenic factors. In addition, C9 disrupted capillary-like networks and newly formed vessels in vitro and rapidly decreased perfusion of neovasculature in vivo. Endothelial cell contraction and membrane blebbing induced by C9 in neovasculature was dependent on the Rho/Rho kinase pathway. CONCLUSIONS AND IMPLICATIONS Anti-angiogenic and vascular disruption by C9 was associated with changes in morphology and function of endothelial cells, involving the Raf-MEK-ERK and Rho/Rho kinase signalling pathways. These findings strongly suggest that C9 is a new microtubule-binding agent that could effectively target tumour vasculature.
Collapse
Affiliation(s)
- Xuan Ren
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | |
Collapse
|
26
|
Mooney CJ, Nagaiah G, Fu P, Wasman JK, Cooney MM, Savvides PS, Bokar JA, Dowlati A, Wang D, Agarwala SS, Flick SM, Hartman PH, Ortiz JD, Lavertu PN, Remick SC. A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid 2009; 19:233-40. [PMID: 19265494 PMCID: PMC2913806 DOI: 10.1089/thy.2008.0321] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fosbretabulin is a novel vascular-disrupting agent that has antitumor activity against anaplastic thyroid cancer (ATC) cell lines, xenografts, and demonstrable efficacy in a phase I trial. This phase II study determined the efficacy and safety of fosbretabulin in patients with advanced ATC and whether fosbretabulin altered the natural history of ATC by virtue of doubling the median survival. A secondary aim evaluated the prognostic value of serum soluble intracellular adhesion molecule-1 (sICAM). METHODS Twenty-six patients received fosbretabulin 45 mg/m(2) as a 10-minute intravenous infusion on days 1, 8, and 15 of a 28-day cycle. sICAM levels were obtained at baseline, over the first two cycles, and end of therapy. Treatment was continued until disease progression. RESULTS Fosbretabulin was well tolerated; grade 3 toxicity was observed in nine patients (35%), and grade 4 toxicity in one (4%). QTc prolongation delayed treatment in four causing one to stop treatment. Median survival was 4.7 months with 34% and 23% alive at 6 and 12 months, respectively. Median duration of stable disease in seven patients was 12.3 months (range, 4.4-37.9 months). Baseline serum sICAM levels were measured in 24 patients with a median 253.5 ng/mL. There was a significant difference in event-free survival among tertiles of baseline sICAM levels (p < 0.009). CONCLUSIONS There were no objective responses seen with single-agent fosbretabulin as administered in this trial, and we did not observe a doubling of survival as our primary endpoint. This is among the largest prospective trials ever conducted for ATC. Fosbretabulin has an acceptable safety profile in patients with advanced ATC, and one-third survived more than 6 months. Despite a small sample size, low baseline sICAM levels were predictive of event-free survival. Further prospective validation of sICAM as a therapeutic biomarker and exploring combination regimens with fosbretabulin are warranted.
Collapse
Affiliation(s)
- Colin J. Mooney
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Govardhanan Nagaiah
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Pingfu Fu
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Biostatistics and Epidemiology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Jay K. Wasman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew M. Cooney
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Panos S. Savvides
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Joseph A. Bokar
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Afshin Dowlati
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ding Wang
- Karmanos Cancer Institute (formerly Josephine Ford Cancer Center), Detroit, Michigan
| | - Sanjiv S. Agarwala
- St. Luke's Hospital and Health Network, Bethlehem, Pennsylvania (formerly University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania)
| | - Susan M. Flick
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Paul H. Hartman
- Translational Research Shared Resource, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Jose D. Ortiz
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Pierre N. Lavertu
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Otolaryngology and Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Scot C. Remick
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
27
|
Dalal S, Burchill SA. Preclinical evaluation of vascular-disrupting agents in Ewing's sarcoma family of tumours. Eur J Cancer 2009; 45:713-22. [PMID: 19136249 DOI: 10.1016/j.ejca.2008.11.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 11/21/2008] [Accepted: 11/24/2008] [Indexed: 02/08/2023]
Abstract
The effects of the tubulin-binding vascular-disrupting agents (VDAs), combretastatin A4 phosphate (CA4P), OXi4503/CA1P and OXi8007, in subcutaneous mouse models of the Ewing's sarcoma family of tumours (ESFTs) have been investigated alone and in combination with doxorubicin. Delay in subcutaneous tumour growth was observed following treatment of mice with multiple doses of OXi4503/CA1P but not with CA4P or OXi8007. A single dose of OXi4503/CA1P caused complete shutdown of vasculature by 24h and extensive haemorrhagic necrosis by 48h. However, a viable rim of proliferating cells remained, which repopulated the tumour within 10 days following the withdrawal of treatment. Combined treatment with doxorubicin 1h prior to administration of OXi4503/CA1P enhanced the effects of OXi4503/CA1P causing a synergistic delay in tumour growth (p<0.001). This study demonstrates that OXi4503/CA1P is a potent VDA in ESFT and in combination with conventional cytotoxic agents represents a promising treatment strategy for this tumour group.
Collapse
Affiliation(s)
- Surita Dalal
- Candlelighter's Children's Cancer Research Group, Cancer Research UK Clinical Centre, Leeds Institute of Molecular Medicine, St. James's University Hospital, Beckett Street, Leeds, West Yorkshire LS9 7TF, UK.
| | | |
Collapse
|
28
|
Tozer GM, Akerman S, Cross NA, Barber PR, Björndahl MA, Greco O, Harris S, Hill SA, Honess DJ, Ireson CR, Pettyjohn KL, Prise VE, Reyes-Aldasoro CC, Ruhrberg C, Shima DT, Kanthou C. Blood Vessel Maturation and Response to Vascular-Disrupting Therapy in Single Vascular Endothelial Growth Factor-A Isoform–Producing Tumors. Cancer Res 2008; 68:2301-11. [DOI: 10.1158/0008-5472.can-07-2011] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Horsman MR, Siemann DW. Pathophysiologic Effects of Vascular-Targeting Agents and the Implications for Combination with Conventional Therapies. Cancer Res 2006; 66:11520-39. [PMID: 17178843 DOI: 10.1158/0008-5472.can-06-2848] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A functional vascular supply is critical for the continued growth and development of solid tumors. It also plays a major role in metastatic spread of tumor cells. This importance has led to the concept of targeting the vasculature of the tumor as a form of cancer therapy. Two major types of vascular-targeting agent (VTA) have now emerged: those that prevent the angiogenic development of the neovasculature of the tumor and those that specifically damage the already established tumor vascular supply. When used alone neither approach readily leads to tumor control, and so, for VTAs to be most successful in the clinic they will need to be combined with more conventional therapies. However, by affecting the tumor vascular supply, these VTAs should induce pathophysiologic changes in variables, such as blood flow, pH, and oxygenation. Such changes could have negative or positive influences on the tumor response to more conventional therapies. This review aims to discuss the pathophysiologic changes induced by VTAs and the implications of these effects on the potential use of VTAs in combined modality therapy.
Collapse
Affiliation(s)
- Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | | |
Collapse
|
30
|
Thoeny HC, De Keyzer F, Chen F, Vandecaveye V, Verbeken EK, Ahmed B, Sun X, Ni Y, Bosmans H, Hermans R, van Oosterom A, Marchal G, Landuyt W. Diffusion-weighted magnetic resonance imaging allows noninvasive in vivo monitoring of the effects of combretastatin a-4 phosphate after repeated administration. Neoplasia 2005; 7:779-87. [PMID: 16207480 PMCID: PMC1501887 DOI: 10.1593/neo.04748] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Revised: 04/06/2005] [Accepted: 05/09/2005] [Indexed: 12/17/2022]
Abstract
The noninvasive assessment of anticancer treatment efficacy is very important for the improvement of therapeutic window. The purpose of the present study was to evaluate the antitumoral effects of the vascular targeting agent, combretastatin A-4 phosphate (CA-4-P), at selected time points after repeated intraperitoneal drug administrations (25 mg/kg), using diffusion-weighted magnetic resonance imaging (DW-MRI). The experiments were performed during an overall follow-up period of 3 weeks on WAG/Rij rats with subcutaneously growing rhabdomyosarcomas. Each animal served as its own baseline. The DW-MRI studies were quantified by calculating the apparent diffusion coefficient (ADC) for different low and high b-values to separate the effects on tumor vasculature and cellular integrity. The changes in ADC as well as the extent of necrosis development (proportional to the tumor volume), measured on the MR images, were of comparable magnitude after each treatment. All ADC values showed a significant decrease at 6 hours, followed by a significant increase at 2 days for various CA-4-P administrations. DW-MRI allowed us to monitor both reduction in perfusion and changes in the extent of tumor necrosis after CA-4-P injection. Repeated CA-4-P administration retains efficacy in rat rhabdomyosarcomas, with similar findings after each drug administration.
Collapse
Affiliation(s)
- Harriet C Thoeny
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
- Department of Diagnostic, Interventional, and Pediatric Radiology, University Hospital of Bern, Freiburgstrasse 10, Bern CH-3010, Switzerland
| | - Frederik De Keyzer
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Feng Chen
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Vincent Vandecaveye
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Erik K Verbeken
- Department of Pathology, University Hospitals Leuven, Kapucijnenvoer 33, Leuven B-3000, Belgium
| | - Bisan Ahmed
- Laboratory of Experimental Radiobiology/LEO, University Hospitals Leuven (KULeuven), Herestraat 49, Leuven B-3000, Belgium
| | - Xihe Sun
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Yicheng Ni
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Hilde Bosmans
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Robert Hermans
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Allan van Oosterom
- Department of Oncology, University Hospitals Leuven, Kapucijnenvoer 33, Leuven B-3000, Belgium
| | - Guy Marchal
- Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Willy Landuyt
- Laboratory of Experimental Radiobiology/LEO, University Hospitals Leuven (KULeuven), Herestraat 49, Leuven B-3000, Belgium
| |
Collapse
|
31
|
Pattillo CB, Sari-Sarraf F, Nallamothu R, Moore BM, Wood GC, Kiani MF. Targeting of the Antivascular Drug Combretastatin to Irradiated Tumors Results in Tumor Growth Delay. Pharm Res 2005; 22:1117-20. [PMID: 16028012 DOI: 10.1007/s11095-005-5646-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Accepted: 04/25/2005] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of the study is to evaluate the effects of targeting the antivascular drug combretastatin to irradiated mouse melanomas. METHODS Combretastatin was incorporated into liposomes with surfaces modified by the addition of cyclo(Arg-Gly-Asp-D-Phe-Cys) (RGD) to create an immunoliposome (IL). This addition of RGD allows the liposome to be preferentially targeted to alphavbeta3, an integrin up-regulated in the vasculature of irradiated tumors. C57BL mice bearing a transplanted B16-F10 melanoma were randomly assigned to one of the following treatment groups: untreated, a single dose of 5-Gy radiation (IR), IL (14.5 mg/kg of combretastatin), 5-Gy radiation plus IL, and a systemic administration of free drug (81.0 mg/kg of combretastatin). RESULTS In this transplanted tumor model, there was no significant increase in the volume of the IL + IR (5 Gy) treated tumors during the initial 6 days posttreatment; all other treatment groups exhibited exponential growth curves after day 3. The IL + IR (5 Gy) treatment resulted in a 5.1-day tumor growth delay compared to untreated controls. CONCLUSIONS These findings indicate that preferential targeting of antivascular drugs to irradiated tumors results in significant tumor growth delay.
Collapse
Affiliation(s)
- Christopher B Pattillo
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
While a number of valid molecular targets have been discovered for tumours over the past decade, finding an effective way of delivering therapeutic genes specifically to tumours has proved more problematic. A variety of viral and non-viral delivery vehicles have been developed and applied in anti-cancer gene therapies. However, these suffer from either inefficient and/or short-lived gene transfer to target cells, instability in the bloodstream and inadequate tumour targeting. Recently, various types of non-pathogenic obligate anaerobic and facultative anaerobic bacteria have been shown to infiltrate and selectively replicate within solid tumours when delivered systemically. This has prompted the development of cancer gene therapy protocols that use such bacteria as gene delivery vehicles. Here, we review the evidence for the success of these in pre-clinical models and clinical trials, as single modality treatments and in combination with conventional cancer therapies.
Collapse
Affiliation(s)
- Rachel M Ryan
- Tumour Targeting Group, Division of Genomic Medicine, University of Sheffield, UK
| | | | | |
Collapse
|
33
|
van den Berg H. Biology and therapy of malignant solid tumors in childhood. ACTA ACUST UNITED AC 2005; 22:643-76. [PMID: 16110632 DOI: 10.1016/s0921-4410(04)22028-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Affiliation(s)
- Henk van den Berg
- Department of Paediatric Oncology, Emma Children's Hospital, Academic Medical Centre, University of Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Abstract
Vascular targeting agents (VTAs) for the treatment of cancer are designed to cause a rapid and selective shutdown of the blood vessels of tumors. Unlike antiangiogenic drugs that inhibit the formation of new vessels, VTAs occlude the pre-existing blood vessels of tumors to cause tumor cell death from ischemia and extensive hemorrhagic necrosis. Tumor selectivity is conferred by differences in the pathophysiology of tumor versus normal tissue vessels (e.g., increased proliferation and fragility, and up-regulated proteins). VTAs can kill indirectly the tumor cells that are resistant to conventional antiproliferative cancer therapies, i.e., cells in areas distant from blood vessels where drug penetration is poor, and hypoxia can lead to radiation and drug resistance. VTAs are expected to show the greatest therapeutic benefit as part of combined modality regimens. Preclinical studies have shown VTA-induced enhancement of the effects of conventional chemotherapeutic agents, radiation, hyperthermia, radioimmunotherapy, and antiangiogenic agents. There are broadly two types of VTAs, small molecules and ligand-based, which are grouped together, because they both cause acute vascular shutdown in tumors leading to massive necrosis. The small molecules include the microtubulin destabilizing drugs, combretastatin A-4 disodium phosphate, ZD6126, AVE8062, and Oxi 4503, and the flavonoid, DMXAA. Ligand-based VTAs use antibodies, peptides, or growth factors that bind selectively to tumor versus normal vessels to target tumors with agents that occlude blood vessels. The ligand-based VTAs include fusion proteins (e.g., vascular endothelial growth factor linked to the plant toxin gelonin), immunotoxins (e.g., monoclonal antibodies to endoglin conjugated to ricin A), antibodies linked to cytokines, liposomally encapsulated drugs, and gene therapy approaches. Combretastatin A-4 disodium phosphate, ZD6126, AVE8062, and DMXAA are undergoing clinical evaluation. Phase I monotherapy studies have shown that the agents are tolerated with some demonstration of single agent efficacy. Because efficacy is expected when the agents are used with conventional chemotherapeutic drugs or radiation, the results of Phase II combination studies are eagerly awaited.
Collapse
Affiliation(s)
- Philip E Thorpe
- Department of Pharmacology and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| |
Collapse
|
35
|
Maxim PG, Carson JJL, Ning S, Knox SJ, Boyer AL, Hsu CP, Benaron DA, Walleczek J. Enhanced Effectiveness of Radiochemotherapy with Tirapazamine by Local Application of Electric Pulses to Tumors. Radiat Res 2004; 162:185-93. [PMID: 15387146 DOI: 10.1667/rr3200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Tumor hypoxia is associated with resistance to radiotherapy and anticancer chemotherapy. However, it can be exploited to therapeutic advantage by concomitantly using hypoxic cytotoxins, such as tirapazamine (TPZ). Tumor electroporation offers the means to further increase tumor hypoxia by temporarily reducing tumor blood flow and therefore increase the cytotoxicity of TPZ. The primary objective of this work was to determine whether electric pulses combined with TPZ and radiotherapy (electroradiochemotherapy) was more efficacious than radiochemotherapy (TPZ + radiation). In these studies using the SCCVII tumor model in C3H mice, electroradiochemotherapy produced up to sixfold more tumor growth delay (TGD) than TPZ + radiation. In these studies, (1) large tumors (280 +/- 15 mm3) responded better to electroradiochemotherapy than small tumors (110 +/- 10 mm3), (2) TGD correlated linearly with tumor volume at the time of electroradiochemotherapy, (3) electric pulses induced a rapid but reversible reduction in O2 saturation, and (4) the electric field was highest near the periphery of the tumor in a 3D computer model. The findings suggested that electroradiochemotherapy gained its therapeutic advantage over TPZ + radiation by enhancing the cytotoxic action of TPZ through reduced tumor oxygenation. The greater antitumor effect achieved in large tumors may be related to tumor morphology and the electric-field distribution. These results suggest that electro-pulsation of large solid tumors may be of benefit to patients treated with radiation in combination with agents that kill hypoxic cells.
Collapse
Affiliation(s)
- Peter G Maxim
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Combretastatin A4 phosphate (CA4P) is a water-soluble prodrug of combretastatin A4 (CA4). The vascular targeting agent CA4 is a microtubule depolymerizing agent. The mechanism of action of the drug is thought to involve the binding of CA4 to tubulin leading to cytoskeletal and then morphological changes in endothelial cells. These changes increase vascular permeability and disrupt tumor blood flow. In experimental tumors, anti-vascular effects are seen within minutes of drug administration and rapidly lead to extensive ischemic necrosis in areas that are often resistant to conventional anti-cancer treatments. Following single-dose administration a viable tumor rim typically remains from which tumor regrowth occurs. When given in combination with therapies targeted at the proliferating viable rim, enhanced tumor responses are seen and in some cases cures. Results from the first clinical trials have shown that CA4P monotherapy is safe and reduces tumor blood flow. There has been some promising demonstration of efficacy. CA4P in combination with cisplatin is also safe. Functional imaging studies have been used to aid the selection of doses for phase II trials. Both dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and positron emission tomography can measure the anti-vascular effects of CA4P in humans. This review describes the background to the development of CA4P, its proposed mechanism of action, the results from the first clinical trials with CA4P and the role of imaging techniques in its clinical development.
Collapse
Affiliation(s)
- Catharine M L West
- Academic Department of Radiation Oncology and Manchester Molecular Imaging Centre, University of Manchester, Christie NHS Trust Hospital, Wilmslow Road, Manchester M20 4BX, UK
| | | |
Collapse
|
37
|
Wildiers H, Ahmed B, Guetens G, De Boeck G, de Bruijn EA, Landuyt W, van Oosterom AT. Combretastatin A-4 phosphate enhances CPT-11 activity independently of the administration sequence. Eur J Cancer 2004; 40:284-90. [PMID: 14728944 DOI: 10.1016/j.ejca.2003.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We evaluated the effect of different intervals and sequences of the vascular targeting agent combretastatin A-4 disodium phosphate (CA4DP) and CPT-11 administration on tumour growth delay and intratumoral uptake of CPT-11 using a syngeneic rhabdomyosarcoma tumour model. Irrespective of the administration sequence, the combination of CA4DP and CPT-11 significantly increases tumour growth delay in comparison with both drugs alone (P<0.001). Intratumoral CPT-11 concentration generally decreased (up to 5-fold) in the combination groups, while SN-38, the active metabolite of CPT-11, increased up to 9-fold. However, the increased amount of intratumoral SN-38 trapping after CA4DP injection did not correlate with the observed tumour growth delay. In conclusion, CA4DP significantly enhances the antitumour effect of CPT-11, which is not greatly influenced by the administration sequence, and which lacks a correlation with the intratumoral trapping of CPT-11 or SN-38. Mechanisms other than trapping are likely to be involved in the chemosensitising capacity of CA4DP.
Collapse
Affiliation(s)
- H Wildiers
- Laboratory of Experimental Oncology, University Hospital Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
38
|
McIntyre DJO, Robinson SP, Howe FA, Griffiths JR, Ryan AJ, Blakey DC, Peers IS, Waterton JC. Single dose of the antivascular agent, ZD6126 (N-acetylcolchinol-O-phosphate), reduces perfusion for at least 96 hours in the GH3 prolactinoma rat tumor model. Neoplasia 2004; 6:150-7. [PMID: 15140404 PMCID: PMC1502085 DOI: 10.1593/neo.03247] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2003] [Revised: 10/16/2003] [Accepted: 10/22/2003] [Indexed: 11/18/2022]
Abstract
Tumor vasculature is an attractive therapeutic target as it differs structurally from normal vasculature, and the destruction of a single vessel can lead to the death of many tumor cells. The effects of antivascular drugs are frequently short term, with regrowth beginning less than 24 hours posttreatment. This study investigated the duration of the response to the vascular targeting agent, ZD6126, of the GH3 prolactinoma, in which efficacy and dose-response have previously been demonstrated. GH3 prolactinomas were grown in the flanks of eight Wistar Furth rats. All animals were treated with 50 mg/kg ZD6126. The tumors were examined with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) 24 hours pretreatment and posttreatment, and at a single time between 48 and 96 hours posttreatment. No evidence of recovery of perfusion was observed even at the longest (96-hour) time point. Involvement of a statistician at the project planning stage and the use of DCE-MRI, which permits noninvasive quantitation of parameters related to blood flow in intact animals, allowed this highly significant result to be obtained using only eight rats.
Collapse
Affiliation(s)
- Dominick J O McIntyre
- Department of Basic Medical Sciences, St. George's Hospital Medical School, Tooting, London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Sheng Y, Hua J, Pinney KG, Garner CM, Kane RR, Prezioso JA, Chaplin DJ, Edvardsen K. Combretastatin family member OXI4503 induces tumor vascular collapse through the induction of endothelial apoptosis. Int J Cancer 2004; 111:604-10. [PMID: 15239140 DOI: 10.1002/ijc.20297] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mechanism of tumor cell killing by OXI4503 was investigated by studying vascular functional and morphological changes post drug administration. SCID mice bearing MHEC5-T hemangioendothelioma were given a single dose of OXI4503 at 100 mg/kg. Tumor blood flow, measured by microsphere fluorescence, was reduced by 50% at 1 hr, and reached a maximum level 6-24 hr post drug treatment. Tumor vascular permeability, measured by Evan's blue and hemoglobin, increased significantly from 3 hr and peaked at 18 hr. The elevated tumor vessel permeability was accompanied by an increase in vascular endothelial growth factor (VEGF) from 1 hr post drug treatment. Immunohistochemical staining for CD31 and laminin showed that tumor blood vessels were affected as early as 3 hr but more prominent from 6 hr. From 12 hr, the vessel structure was completely destroyed. Histopathological and double immunohistochemical staining showed morphological change and induction of apoptosis in endothelial cells at 1-3 hr, followed by tumor cell necrosis from 6-72 hr. There were no statistically significant changes of Evan's blue and hemoglobin contents in liver tissue over the time course. These results suggest that OXI4503 selectively targets tumor blood vessels, and induces blood flow shutdown while it enhances tumor blood vessel permeability. The early induction of endothelial cell apoptosis leads to functional changes of tumor blood vessels and finally to the collapse of tumor vasculature, resulting in massive tumor cell necrosis. The time course of the tumor vascular response observed with OXI4503 treatment supports this drug for development as a stand alone therapy, and also lends support for the use of the drug in combination with other cancer therapies.
Collapse
Affiliation(s)
- Yezhou Sheng
- Department of Cell and Molecular Biology, Section for Tumor Immunology, University of Lund, Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Horsman MR, Murata R. Vascular targeting effects of ZD6126 in a C3H mouse mammary carcinoma and the enhancement of radiation response. Int J Radiat Oncol Biol Phys 2003; 57:1047-55. [PMID: 14575836 DOI: 10.1016/s0360-3016(03)00769-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE The aim of this study was to investigate the pathophysiologic effects induced by the novel vascular targeting agent ZD6126 in a C3H mouse mammary carcinoma and to evaluate the agent's ability to inhibit tumor growth either when given alone or in combination with radiation. METHODS AND MATERIALS A C3H mammary carcinoma grown in the right rear foot of female CDF1 mice was treated when at 200 mm(3) in size. ZD6126 was dissolved in saline and injected intraperitoneally. Blood perfusion was measured using the RbCl extraction procedure, tumor oxygen (pO(2)) status was assessed with the Eppendorf electrode, and tumor necrosis was estimated from histologic sections. Radiation (240-kV X-rays) was locally administered to tumors of restrained nonanesthetized mice, and response was assessed using a tumor growth assay. RESULTS ZD6126 induced a significant dose- and time-dependent decrease in tumor perfusion, reaching a maximal 70% reduction around 3 h after injecting 150-300 mg/kg. However, full recovery was seen within 6 h. A 200 mg/kg dose significantly decreased tumor oxygenation status at 3 h (median pO(2) decreased from 7 to 3 mm Hg; % pO(2) values <or=2.5 mm Hg increased from 30% to 55%) and by 24 h had significantly increased necrotic fraction from 14.5% to 25.2%. This ZD6126 dose resulted in a small, yet significant, 1.4 days inhibition of tumor growth when given alone. It also enhanced the tumor response to radiation, giving rise to a significant 1.3-fold increase in the slope of the radiation dose-response curve. Of the normal tissues, only muscle (at 3 h) and spleen (at 6 h) showed any significant reduction in perfusion after injecting 200 mg/kg, but these transient decreases were only 32% and 49%, respectively. CONCLUSIONS Our preclinical studies clearly demonstrate a tumor-specific reduction in blood perfusion by ZD6126. Although these changes were transient, they were sufficient to increase tumor necrosis, inhibit tumor growth, and enhance radiation response.
Collapse
Affiliation(s)
- Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | | |
Collapse
|
41
|
Hori K, Saito S. Microvascular mechanisms by which the combretastatin A-4 derivative AC7700 (AVE8062) induces tumour blood flow stasis. Br J Cancer 2003; 89:1334-44. [PMID: 14520469 PMCID: PMC2394288 DOI: 10.1038/sj.bjc.6601261] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We previously reported that a novel combretastatin A-4 derivative, AC7700, has remarkable antitumour effects because of an irreversible stasis of tumour blood flow (TBF) and subsequent loss of nutrient supply to tumour tissue. Since early 2002, under the new designation AVE8062, AC7700 has undergone clinical trials in Europe and the US. Questions remain, however, concerning how AC7700 blocks TBF and why the TBF stasis does not recover. In this study, using a rat tumour LY80, a variant of Yoshida sarcoma, we examined whether TBF cessation after AC7700 administration is due to a direct action of the agent on tumour blood vessels. We constructed electrodes that can drop a small quantity of the drug solution directly at the site of blood flow measurement and inserted them subcutaneously and into the tumour. We compared the blood flow responses of normal vessels and tumour vessels after administration of 10-μl doses of various concentrations (0.2, 1, 10, and 50 mg ml−1) of the AC7700 solution. In addition, we assessed TBF stasis after i.v. and intra-arterial 10 mg kg−1 AC7700 administration in an LY80-induced kidney tumour. To determine why the TBF stasis is irreversible, we observed AC7700-induced changes in host arterioles and the tumour vascular network of the Sato lung carcinoma using a vital microscopic rat transparent chamber. Since an increase in tumour interstitial fluid pressure brings about a decrease in TBF, we also measured 10 mg kg−1 AC7700-induced changes in this pressure. The sensitivity of the blood flow response after intratumoral application of AC7700 was markedly higher in normal vessels relative to tumour vessels. Intra-arterial administration of AC7700 did not have stronger effects on TBF stasis than did i.v. administration. Intravital microscopy showed that AC7700 induced a powerful and long-lasting constriction of host arterioles, so that complete stasis of blood flow occurred in downstream vessels, which supplied blood to tumours. Owing to this stasis, the lumens of numerous tumour vessels narrowed or completely disappeared, and numerous erythrocytes stagnated in drainage vessels of the tumour vascular network. Haemolysis of these erythrocytes occurred after 2–3 h, resulting in complete thrombosis. There was no indication of reperfusion in vessels showing haemolysis. This haemolysis is thought to be the main cause for the irreversibility of TBF stasis. Since the tumour interstitial fluid pressure decreased after i.v. AC7700 administration, the possibility of stasis of TBF being caused by tumour vascular compression was excluded. All these results strongly suggest that the main target of AC7700 is host arterioles and that the stasis of TBF induced by AC7700 is not triggered by a direct action of the drug on tumour vessels.
Collapse
Affiliation(s)
- K Hori
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Sendai 980-8575, Japan.
| | | |
Collapse
|
42
|
Lefesvre P, Attema J, van Bekkum D. Adenoviral gene transfer of angiostatic ATF-BPTI inhibits tumour growth. BMC Cancer 2002; 2:17. [PMID: 12147175 PMCID: PMC122087 DOI: 10.1186/1471-2407-2-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2002] [Accepted: 07/29/2002] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The outgrowth of new vessels--angiogenesis--in the tumour mass is considered to be a limiting factor of tumour growth. To inhibit the matrix lysis that is part of the tumour angiogenesis, we employed the chimeric protein mhATF-BPTI, composed of the receptor binding part of the urokinase (ATF) linked to an inhibitor of plasmin (BPTI). METHODS For delivery, recombinant adenovirus encoding the transgene of interest was injected intravenously or locally into the tumour. The anti tumour effect of this compound was compared to that of human endostatin and of mhATF alone in two different rat bronchial carcinomas growing either as subcutaneous implants or as metastases. RESULTS Significant inhibition of the tumour growth and decrease of the number of lung metastasis was achieved when the concentration of mhATF-BPTI at the tumour site was above 400 of ng/g tissue. This concentration could be achieved via production by the liver, only if permissive to the recombinant adenovirus. When the tumour cells could be transduced, local delivery of the vector was enough to obtain a response. In the case of metastasis, the capacity of the lung tissue to concentrate the encoded protein was essential to reach the required therapeutic levels. Further, endostatin or mhATF could not reproduce the effects of mhATF-BPTI, at similar concentrations (mhATF) and even at 10-fold higher concentration (endostatin). CONCLUSION The ATF-BPTI was shown to inhibit tumour growth of different rat lung tumours when critical concentration was reached. In these tumour models, endostatin or ATF induce almost no tumour response.
Collapse
MESH Headings
- Adenoviridae/genetics
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/genetics
- Animals
- Aprotinin/administration & dosage
- Aprotinin/genetics
- Aprotinin/pharmacology
- Aprotinin/therapeutic use
- Carcinoma, Bronchogenic/blood supply
- Carcinoma, Bronchogenic/drug therapy
- Carcinoma, Bronchogenic/metabolism
- Carcinoma, Bronchogenic/secondary
- Collagen/pharmacology
- Collagen/therapeutic use
- Drug Delivery Systems/methods
- Endostatins
- Gene Transfer Techniques
- Genetic Vectors/administration & dosage
- Genetic Vectors/genetics
- Genetic Vectors/pharmacology
- Genetic Vectors/therapeutic use
- Injections, Intralesional
- Injections, Intravenous
- Lung Neoplasms/blood supply
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Male
- Neoplasm Transplantation
- Peptide Fragments/pharmacology
- Peptide Fragments/therapeutic use
- Rats
- Rats, Inbred BN
- Rats, Inbred Strains
- Receptors, Cell Surface/administration & dosage
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/therapeutic use
- Receptors, Urokinase Plasminogen Activator
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/therapeutic use
- Serine Proteinase Inhibitors/administration & dosage
- Serine Proteinase Inhibitors/genetics
- Serine Proteinase Inhibitors/pharmacology
- Serine Proteinase Inhibitors/therapeutic use
- Urokinase-Type Plasminogen Activator
Collapse
Affiliation(s)
| | - Joline Attema
- Crucell BV, PO BOX 2048, 2301CA, Leiden The Netherlands
| | | |
Collapse
|
43
|
Hori K, Saito S, Kubota K. A novel combretastatin A-4 derivative, AC7700, strongly stanches tumour blood flow and inhibits growth of tumours developing in various tissues and organs. Br J Cancer 2002; 86:1604-14. [PMID: 12085211 PMCID: PMC2746587 DOI: 10.1038/sj.bjc.6600296] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2001] [Revised: 03/04/2002] [Accepted: 03/12/2002] [Indexed: 11/09/2022] Open
Abstract
In a previous study, we used subcutaneous LY80 tumours (a subline of Yoshida sarcoma), Sato lung carcinoma, and methylcholanthrene-induced primary tumours, to demonstrate that a novel water-soluble combretastatin A-4 derivative, AC7700, abruptly and irreversibly stopped tumour blood flow. As a result of this interrupted supply of nutrients, extensive necrosis was induced within the tumour. In the present study, we investigated whether AC7700 acts in the same way against solid tumours growing in the liver, stomach, kidney, muscle, and lymph nodes. Tumour blood flow and the change in tumour blood flow induced by AC7700 were measured by the hydrogen clearance method. In a model of cancer chemotherapy against metastases, LY80 cells (2x10(6)) were injected into the lateral tail vein, and AC7700 at 10 mg x kg(-1) was injected i.v. five times at intervals of 2 days, starting on day 7 after tumour cell injection. The number and size of tumours were compared with those in the control group. The change in tumour blood flow and the therapeutic effect of AC7700 on microtumours were observed directly by using Sato lung carcinoma implanted in a rat transparent chamber. AC7700 caused a marked decrease in the tumour blood flow of all LY80 tumours developing in various tissues and organs and growth of all tumours including lymph node metastases and microtumours was inhibited. In every tumour, tumour blood flow began to decrease immediately after AC7700 administration and reached a minimum at approximately 30 min after injection. In many tumour capillaries, blood flow completely stopped within 3 min after AC7700 administration. These results demonstrate that AC7700 is effective for tumours growing in various tissues and organs and for metastases. We conclude that tumour blood flow stanching induced by AC7700 may become an effective therapeutic strategy for all cancers, including refractory cancers because the therapeutic effect is independent of tumour site and specific type of cancer.
Collapse
Affiliation(s)
- K Hori
- Department of Vascular Biology, Division of Cancer Control, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | |
Collapse
|
44
|
Eikesdal HP, Landuyt W, Dahl O. The influence of combretastatin A-4 and vinblastine on interstitial fluid pressure in BT4An rat gliomas. Cancer Lett 2002; 178:209-17. [PMID: 11867206 DOI: 10.1016/s0304-3835(01)00835-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The influence of combretastatin A-4 disodium phosphate (CA-4, 50mg/kg intraperitoneally (i.p.)) and vinblastine (2mg/kg i.p.) on interstitial fluid pressure (IFP) was assessed in BT4An rat gliomas implanted subcutaneously in the neck. Furthermore the growth inhibitory effect of vinblastine and the distribution of fluorescence-conjugated vinblastine (BODIPY-vinblastine) were investigated. Tumors at different volumes were compared. Whereas CA-4 had no major influence on IFP, independent of tumor size, vinblastine increased the IFP in neoplasms above 8 cm(3) (P=0.03). Vinblastine yielded a significant tumor response only in tumors below 2.1 cm(3) (P=0.03). The distribution of BODIPY-vinblastine was heterogeneous and comparable despite tumor volume differences. We conclude that the influence of vinblastine on IFP is more pronounced than that of CA-4 in BT4An neck tumors, and that vinblastine may reduce subsequent drug delivery to solid tumors by increasing the IFP.
Collapse
Affiliation(s)
- Hans Petter Eikesdal
- Department of Oncology, University of Bergen, Haukeland University Hospital, 5021 Bergen, Norway.
| | | | | |
Collapse
|
45
|
Basha G, Landuyt W, Fowler J, Vordermark D, Haustermans K, Geboes K, Van den Bogaert W, Yap SH, Lambin P, Penninckx F. An experimental evaluation of three preoperative radiation regimens for resectable rectal cancer. Ann Surg Oncol 2002; 9:292-7. [PMID: 11923137 DOI: 10.1007/bf02573068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND We investigated the degree of tumor cell killing after radiotherapy regimens commonly used in clinical practice in comparison with an accelerated schedule. METHODS Mtln3 mammary adenocarcinoma tumor cells were inoculated subcutaneously in the hind leg of syngeneic Fischer 344 rats. Tumors were irradiated with 5 x 5 Gy in 5 days, 10 x 3 Gy over 10 days, or 5 x (2 x 3) Gy in 5 days. After excision of the irradiated tumors, the dye exclusion, a tetrazolium-based colorimetric and the clonogenic assays were used to determine tumor cell viability and surviving fractions. RESULTS Estimated potential doubling time values indicate a rapid proliferation capacity, comparable with potential doubling time values in human rectal cancer. The dye exclusion and clonogenic assays revealed a significantly higher degree of cell killing after the hypofractionated and the accelerated regimens of, respectively, 5 x 5 Gy and 5 x (2 x 3) Gy over 5 days compared with 10 x 3 Gy over 10 days. CONCLUSIONS A shorter treatment time offered the best therapeutic efficacy. The schedule involving two daily fractions of 3 Gy over 5 days should be less toxic than 5 x 5 Gy and may therefore provide a therapeutic advantage.
Collapse
Affiliation(s)
- Genc Basha
- Department of Abdominal Surgery, University Hospital Gasthuisberg, Katholieke Universiteit Leuven, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wouters BG, Weppler SA, Koritzinsky M, Landuyt W, Nuyts S, Theys J, Chiu RK, Lambin P. Hypoxia as a target for combined modality treatments. Eur J Cancer 2002; 38:240-57. [PMID: 11803141 DOI: 10.1016/s0959-8049(01)00361-6] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is overwhelming evidence that solid human tumours grow within a unique micro-environment. This environment is characterised by an abnormal vasculature, which leads to an insufficient supply of oxygen and nutrients to the tumour cells. These characteristics of the environment limit the effectiveness of both radiotherapy and chemotherapy. Measurement of the oxygenation status of human tumours has unequivocally demonstrated the importance of this parameter on patient prognosis. Tumour hypoxia has been shown to be an independent prognostic indicator of poor outcome in prostate, head and neck and cervical cancers. Recent laboratory and clinical data have shown that hypoxia is also associated with a more malignant phenotype, affecting genomic stability, apoptosis, angiogenesis and metastasis. Several years ago, scientists realised that the unique properties within the tumour micro-environment could provide the basis for tumour-specific therapies. Efforts that are underway to develop therapies that exploit the tumour micro-environment can be categorised into three groups. The first includes agents that exploit the environmental changes that occur within the micro-environment such as hypoxia and reduced pH. This includes bioreductive drugs that are specifically toxic to hypoxic cells, as well as hypoxia-specific gene delivery systems. The second category includes therapies designed to exploit the unique properties of the tumour vasculature and include both angiogenesis inhibitors and vascular targeting agents. The final category includes agents that exploit the molecular and cellular responses to hypoxia. For example, many genes are induced by hypoxia and promoter elements from these genes can be used for the selective expression of therapeutic proteins in hypoxic tumour cells. An overview of the various properties ascribed to tumour hypoxia and the current efforts underway to exploit hypoxia for improving cancer treatment will be discussed.
Collapse
Affiliation(s)
- B G Wouters
- Department of Experimental Radiation Oncology, UNS 50/ Box 23, azM/University of Maastricht, Postbox 616, 6200 MD Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Eikesdal HP, Bjerkvig R, Raleigh JA, Mella O, Dahl O. Tumor vasculature is targeted by the combination of combretastatin A-4 and hyperthermia. Radiother Oncol 2001; 61:313-20. [PMID: 11731002 DOI: 10.1016/s0167-8140(01)00450-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND PURPOSE Combretastatin A-4 disodium phosphate (CA-4) enhances thermal damage in s.c. BT(4)An rat gliomas. We currently investigated how CA-4 and hyperthermia affect the tumor microenvironment and neovasculature to disclose how the two treatment modalities interact to produce tumor response. METHODS By confocal microscopy and immunostaining for von Willebrand factor, we examined the extent of vascular damage subsequent to CA-4 (50 mg/kg) and hyperthermia (waterbath 44 degrees C, 60 min). The influence on tumor oxygenation was assessed using interstitial pO(2)-probes (Licox system) and by immunostaining for pimonidazole. We examined the direct effect of CA-4 on the tumor cell population by flow cytometry (cell cycle distribution) and immunostaining for beta-tubulin (cytoskeletal damage). RESULTS Whereas slight vascular damage was produced by CA-4 in the BT(4)An tumors, local hyperthermia exhibited moderate anti-vascular activity. In tumors exposed to CA-4 3 h before hyperthermia, massive vascular damage ensued. CA-4 reduced the pO(2) from 36.1 to 17.6 mmHg (P=0.01) in the tumor base, and tumor hypoxia increased slightly in the tumor center (pimonidazole staining). Extensive tumor hypoxia developed subsequent to hyperthermia or combination therapy. Despite a profound influence on beta-tubulin organization in vitro, CA-4 had no significant effect on the cell cycle distribution in vivo. CONCLUSION Our results indicate that the anti-vascular activity exhibited by local hyperthermia can be augmented by previous exposure to CA-4.
Collapse
Affiliation(s)
- H P Eikesdal
- Department of Oncology, University of Bergen, Haukeland University Hospital, 5021 Bergen, Norway
| | | | | | | | | |
Collapse
|
48
|
Eikesdal HP, Bjerkvig R, Dahl O. Vinblastine and hyperthermia target the neovasculature in BT(4)AN rat gliomas: therapeutic implications of the vascular phenotype. Int J Radiat Oncol Biol Phys 2001; 51:535-44. [PMID: 11567830 DOI: 10.1016/s0360-3016(01)01693-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE The antivascular and antitumor activity of vinblastine and hyperthermia at different tumor volumes were examined in the subcutaneous (s.c.) BT(4)An rat glioma model. METHODS AND MATERIALS The influence of vinblastine (3 mg/kg) and hyperthermia (44 degrees C/60 min) on tumor growth was assessed in small (100 mm(3)) and large (200 mm(3)) BT(4)An tumors. To disclose how vinblastine and hyperthermia interacted in the neoplasms, tumor blood flow and the extent of vascular damage, hypoxia, cell proliferation, and apoptosis were assessed after treatment. The content of smooth muscle cells/pericytes in the tumor vasculature was examined in small and large tumors to assess how the vascular phenotype changed during tumor growth. RESULTS In the large tumors, vinblastine reduced the blood flow, but the tumor growth was not affected. The combination of drug and local heating yielded massive vascular damage and a significant tumor response. The small neoplasms had a higher content of smooth muscle cells/pericytes in the vessel walls (host vasculature), and the tumor vasculature displayed a higher resistance to vascular damage than the large neoplasms. Yet, vinblastine alone exhibited a potent antiproliferative activity and induced massive apoptosis in the small tumors, and the drug significantly inhibited tumor growth. The addition of hyperthermia yielded no additional growth delay in the small tumors. CONCLUSION The antivascular properties of vinblastine and hyperthermia can be exploited to facilitate vascular damage in BT(4)An solid tumors with a low content of host vasculature.
Collapse
Affiliation(s)
- H P Eikesdal
- Department of Oncology, Institute of Medicine, University of Bergen, Haukeland Hospital, Bergen, Norway.
| | | | | |
Collapse
|
49
|
Theys J, Landuyt W, Nuyts S, Van Mellaert L, van Oosterom A, Lambin P, Anné J. Specific targeting of cytosine deaminase to solid tumors by engineered Clostridium acetobutylicum. Cancer Gene Ther 2001; 8:294-7. [PMID: 11393282 DOI: 10.1038/sj.cgt.7700303] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The presence of severe hypoxia and necrosis in solid tumors offers the potential to apply an anaerobic bacterial enzyme/prodrug approach in cancer treatment. In this context the apathogenic C. acetobutylicum was genetically engineered to express and secrete E. coli cytosine deaminase (CDase). Considerable levels of functional cytosine deaminase were detected in lysates and supernatants of recombinant C acetobutylicum cultures. After administration of the recombinant Clostridium to rhabdomyosarcoma bearing rats used as a model, cytosine deaminase could be detected at the tumor site. Moreover, following administration of the vascular targeting agent combretastatin A-4 phosphate significantly increased levels of cytosine deaminase were detected at the tumor site as a consequence of enlarged tumor necrosis and subsequently improved growth of C. acetobutylicum. The results provide evidence for the potential application of Clostrisdium-based therapeutic protein transfer to tumors in anticancer therapy.
Collapse
Affiliation(s)
- J Theys
- Laboratory of Bacteriology, Rega Institute for Medical Research, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
50
|
Theys J, Landuyt W, Nuyts S, Van Mellaert L, Bosmans E, Rijnders A, Van Den Bogaert W, van Oosterom A, Anné J, Lambin P. Improvement of Clostridium tumour targeting vectors evaluated in rat rhabdomyosarcomas. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2001; 30:37-41. [PMID: 11172989 DOI: 10.1111/j.1574-695x.2001.tb01547.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous studies have demonstrated the feasibility of using apathogenic clostridia as a promising strategy for hypoxia-specific tumour targeting. The present study shows that the use of the vascular targeting compound combretastatin A-4 phosphate could significantly (P<0.001) increase the number of Clostridium vegetative cells in rat rhabdomyosarcomas with sizes between 0.2 cm(2) and 3 cm(2). Furthermore, this study showed that administration of metronidazole for a 9-day period was sufficient to eliminate systemically administered Clostridium from the tumour. Moreover, previous Clostridium spore administration did not effect tumour colonisation, regardless of the immune response status of the host.
Collapse
Affiliation(s)
- J Theys
- Laboratory of Bacteriology, Rega Institute for Medical Research, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|