1
|
Krolewski JJ, Singh S, Sha K, Jaiswal N, Turowski SG, Pan C, Rich LJ, Seshadri M, Nastiuk KL. TNF Signaling Is Required for Castration-Induced Vascular Damage Preceding Prostate Cancer Regression. Cancers (Basel) 2022; 14:cancers14246020. [PMID: 36551505 PMCID: PMC9775958 DOI: 10.3390/cancers14246020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The mainstay treatment for locally advanced, recurrent, or metastatic prostate cancer (PrCa) is androgen deprivation therapy (ADT). ADT causes prostate cancers to shrink in volume, or regress, by inducing epithelial tumor cell apoptosis. In normal, non-neoplastic murine prostate, androgen deprivation via castration induces prostate gland regression that is dependent on TNF signaling. In addition to this direct mechanism of action, castration has also been implicated in an indirect mechanism of prostate epithelial cell death, which has been described as vascular regression. The initiating event is endothelial cell apoptosis and/or increased vascular permeability. This subsequently leads to reduced blood flow and perfusion, and then hypoxia, which may enhance epithelial cell apoptosis. Castration-induced vascular regression has been observed in both normal and neoplastic prostates. We used photoacoustic, power Doppler, and contrast-enhanced ultrasound imaging, and CD31 immunohistochemical staining of the microvasculature to assess vascular integrity in the period immediately following castration, enabling us to test the role of TNF signaling in vascular regression. In two mouse models of androgen-responsive prostate cancer, TNF signaling blockade using a soluble TNFR2 ligand trap reversed the functional aspects of vascular regression as well as structural changes in the microvasculature, including reduced vessel wall thickness, cross-sectional area, and vessel perimeter length. These results demonstrate that TNF signaling is required for vascular regression, most likely by inducing endothelial cell apoptosis and increasing vessel permeability. Since TNF is also the critical death receptor ligand for prostate epithelial cells, we propose that TNF is a multi-purpose, comprehensive signal within the prostate cancer microenvironment that mediates prostate cancer regression following androgen deprivation.
Collapse
Affiliation(s)
- John J. Krolewski
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Shalini Singh
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kai Sha
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Neha Jaiswal
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Steven G. Turowski
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chunliu Pan
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Laurie J. Rich
- Laboratory of Translational Imaging, Center for Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mukund Seshadri
- Laboratory of Translational Imaging, Center for Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kent L. Nastiuk
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Correspondence: ; Tel.: +1-716-845-5771
| |
Collapse
|
2
|
F. Antunes I, Haisma HJ, Elsinga PH, Di Gialleonardo V, Waarde AV, Willemsen ATM, Dierckx RA, de Vries EFJ. Induction of β-Glucuronidase Release by Cytostatic Agents in Small Tumors. Mol Pharm 2012; 9:3277-85. [DOI: 10.1021/mp300327w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Inês F. Antunes
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Hidde J. Haisma
- Department of Pharmaceutical
Gene Modulation, University Center for Pharmacy, University of Groningen,
A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Philip H. Elsinga
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Valentina Di Gialleonardo
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Antoon T. M. Willemsen
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Rudi A. Dierckx
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Erik F. J. de Vries
- Department of Nuclear Medicine
and Molecular Imaging, University Medical Center Groningen, University
of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
3
|
Crescenzi M, Persano L, Esposito G, Zulato E, Borsi L, Balza E, Ruol A, Ancona E, Indraccolo S, Amadori A. Vandetanib Improves Anti-Tumor Effects of L19mTNFα in Xenograft Models of Esophageal Cancer. Clin Cancer Res 2010; 17:447-58. [DOI: 10.1158/1078-0432.ccr-10-1420] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
4
|
|
5
|
Hypoxia and the metabolic phenotype of prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:1433-43. [DOI: 10.1016/j.bbabio.2009.06.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 05/21/2009] [Accepted: 06/07/2009] [Indexed: 12/27/2022]
|
6
|
Menon C, Bauer TW, Kelley ST, Raz DJ, Bleier JI, Patel K, Steele K, Prabakaran I, Shifrin A, Buerk DG, Sehgal CM, Fraker DL. Tumoricidal activity of high-dose tumor necrosis factor-alpha is mediated by macrophage-derived nitric oxide burst and permanent blood flow shutdown. Int J Cancer 2008; 123:464-475. [PMID: 18449880 DOI: 10.1002/ijc.23499] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study investigates the role of tumor nitric oxide (NO) and vascular regulation in tumor ulceration following high-dose tumor necrosis factor-alpha (TNF) treatment. Using TNF-responsive (MethA) and nonresponsive (LL2) mouse tumors, tumor NO concentration was measured with an electrochemical sensor and tumor blood flow by Doppler ultrasound. Mice were also pretreated with a selective inducible nitric oxide synthase (iNOS) inhibitor, 1400 W. Tumors harvested from TNF-treated mice were cryosectioned and immunostained for murine macrophages, or/and iNOS. MethA tumor-bearing mice were depleted of macrophages. Pre- and post-TNF tumor NO levels were measured continuously, and mice were followed for gross tumor response. In MethA tumors, TNF caused a 96% response rate, and tumor NO concentration doubled. Tumor blood flow decreased to 3% of baseline by 4 hr and was sustained at 24 hr and 10 days post-TNF. Selective NO inhibition with 1400 W blocked NO rise and decreased response rate to 38%. MethA tumors showed tumor infiltration by macrophages post-TNF and the pattern of macrophage immunostaining overlapped with iNOS immunostaining. Depletion of macrophages inhibited tumor NO increase and response to TNF. LL2 tumors had a 0% response rate to TNF and exhibited no change in NO concentration. Blood flow decreased to 2% of baseline by 4 hr, recovered to 56% by 24 hr and increased to 232% by 10 days. LL2 tumors showed no infiltration by macrophages post-TNF. We conclude that TNF causes tumor infiltrating, macrophage-derived iNOS-mediated tumor NO rise and sustained tumor blood flow shutdown, resulting in tumor ulceration in the responsive tumor.
Collapse
Affiliation(s)
| | - Todd W Bauer
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Scott T Kelley
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Dan J Raz
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Joshua I Bleier
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Krina Patel
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Kirsten Steele
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | | | | | - Donald G Buerk
- Department of Physiology, University of Pennsylvania, Philadelphia, PA
| | - Chandra M Sehgal
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Douglas L Fraker
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
7
|
Palanker D, Vankov A, Freyvert Y, Huie P. Pulsed electrical stimulation for control of vasculature: Temporary vasoconstriction and permanent thrombosis. Bioelectromagnetics 2008; 29:100-7. [DOI: 10.1002/bem.20368] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
8
|
Cooney MM, van Heeckeren W, Bhakta S, Ortiz J, Remick SC. Drug insight: vascular disrupting agents and angiogenesis--novel approaches for drug delivery. ACTA ACUST UNITED AC 2007; 3:682-92. [PMID: 17139319 DOI: 10.1038/ncponc0663] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 09/06/2006] [Indexed: 12/13/2022]
Abstract
Vascular disrupting agents (VDAs), or endothelial disrupting agents, attempt to exploit the vascular endothelium that supplies rapidly dividing neoplasms. Unlike antiangiogenesis agents (e.g. the monoclonal antibody bevacizumab; and tyrosine kinase inhibitors sorafenib and sunitinib) that disrupt endothelial cell survival mechanisms and the development of a new tumor blood supply, VDAs are designed to disrupt the already established abnormal vasculature that supports tumors, by targeting their dysmorphic endothelial cells. Tumor vascular endothelium is characterized by its increased permeability, abnormal morphology, disorganized vascular networks, and variable density. VDAs induce rapid shutdown of tumor blood supply, causing subsequent tumor death from hypoxia and nutrient deprivation. The safety profile of this class of compounds is more indicative of agents that are indeed 'vascularly' active. For example, VDAs can cause: acute coronary and other thrombophlebitic syndromes; alterations in blood pressure, heart rate, and ventricular conduction; transient flush and hot flashes; neuropathy; and tumor pain. Despite these cardiovascular concerns some patients have benefited from VDAs in early clinical trials. Further drug development of VDAs must include the combination of these agents with other novel biological agents, cytotoxic chemotherapy, and radiotherapy. Close monitoring of patients receiving VDAs for any cardiovascular toxicity is imperative.
Collapse
Affiliation(s)
- Matthew M Cooney
- University Hospitals of Cleveland, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
| | | | | | | | | |
Collapse
|
9
|
McPhail LD, McIntyre DJO, Ludwig C, Kestell P, Griffiths JR, Kelland LR, Robinson SP. Rat tumor response to the vascular-disrupting agent 5,6-dimethylxanthenone-4-acetic acid as measured by dynamic contrast-enhanced magnetic resonance imaging, plasma 5-hydroxyindoleacetic acid levels, and tumor necrosis. Neoplasia 2006; 8:199-206. [PMID: 16611413 PMCID: PMC1578525 DOI: 10.1593/neo.05739] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The dose-dependent effects of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) on rat GH3 prolactinomas were investigated in vivo. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used to assess tumor blood flow/permeability pretreatment and 24 hours posttreatment with 0, 100, 200, or 350 mg/kg DMXAA. DCE-MRI data were analyzed using K(trans) and the integrated area under the gadolinium time curve (IAUGC) as response biomarkers. High-performance liquid chromatography (HPLC) was used to determine the plasma concentration of the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) following treatment to provide an index of increased vessel permeability and vascular damage. Finally, tumor necrosis was assessed by grading hematoxylin and eosin-stained sections cut from the same tumors investigated by MRI. Both tumor K(trans) and IAUGC were significantly reduced 24 hours posttreatment with 350 mg/kg DMXAA only, with no evidence of dose response. HPLC demonstrated a significant increase in plasma 5-HIAA 24 hours posttreatment with 200 and 350 mg/kg DMXAA. Histologic analysis revealed some evidence of tumor necrosis following treatment with 100 or 200 mg/kg DMXAA, reaching significance with 350 mg/kg DMXAA. The absence of any reduction in K(trans) or IAUGC following treatment with 200 mg/kg, despite a significant increase in 5-HIAA, raises concerns about the utility of established DCE-MRI biomarkers to assess tumor response to DMXAA.
Collapse
Affiliation(s)
- Lesley D McPhail
- Division of Basic Medical Sciences, St. George's, University of London, London, UK.
| | | | | | | | | | | | | |
Collapse
|
10
|
van Laarhoven HWM, Gambarota G, Heerschap A, Lok J, Verhagen I, Corti A, Toma S, Gallo Stampino C, van der Kogel A, Punt CJA. Effects of the tumor vasculature targeting agent NGR-TNF on the tumor microenvironment in murine lymphomas. Invest New Drugs 2006; 24:27-36. [PMID: 16379040 DOI: 10.1007/s10637-005-4540-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
TNF-alpha may improve drug delivery to tumors by alteration of vascular permeability. However, toxicity precludes its systemic administration in patients. NGR-TNF comprises TNF coupled to the peptide CNGRC, which is a ligand for CD13. CD13 is expressed on tumor vasculature. Therefore, to assess the efficacy of NGR-TNF its biological effect on tumor vasculature should be measured rather than its effect on tumor growth. The aim of this study was to assess the effects of a low dose of NGR-TNF (5 ng/kg) on vascular permeability, tumor hypoxia, perfusion and proliferation in lymphoma bearing mice. MRI measurements with blood pool contrast agent showed an increased leakage of the contrast agent from the vasculature in NGR-TNF treated tumors compared with controls (p < 0.05), suggesting NGR-TNF-induced vascular permeability. Immunohistochemical analysis two hours after NGR-TNF treatment showed a decrease in tumor hypoxia (p < 0.1) and an increase in labeling index of the S-phase marker bromodeoxyuridine (p < 0.1), possibly due to an increase in tumor blood flow after NGR-TNF treatment. Although a decrease in tumor hypoxia and an increase in labeling index could have lead to increased tumor growth, in this experiment after one day a decrease in tumor volume was measured. Possibly, the effects on tumor hypoxia and proliferation two hours after treatment are transient and overruled by other, more longlasting effects. For example, the observed increase in vascular permeability may lead to haemoconcentration and increased interstitial pressure, ultimately resulting in an reduction of tumor blood flow and thus a decrease in tumor growth. A beneficial effect of NGR-TNF in combination with other therapeutical agents may therefore critically depend on the sequence and timing of the regimens. Currently, NGR-TNF is being tested in clinical studies.
Collapse
Affiliation(s)
- H W M van Laarhoven
- Department of Medical Oncology, University Medical Centre Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Low-molecular-weight vascular-disrupting agents (VDAs) cause a pronounced shutdown in blood flow to solid tumours, resulting in extensive tumour-cell necrosis, while they leave the blood flow in normal tissues relatively intact. The largest group of VDAs is the tubulin-binding combretastatins, several of which are now being tested in clinical trials. DMXAA (5,6-dimethylxanthenone-4-acetic acid) - one of a structurally distinct group of drugs - is also being tested in clinical trials. A full understanding of the action of these and other VDAs will provide insights into mechanisms that control tumour blood flow and will be the basis for the development of new therapeutic drugs for targeting the established tumour vasculature for therapy.
Collapse
Affiliation(s)
- Gillian M Tozer
- Academic Unit of Surgical Oncology, Division of Clinical Sciences, University of Sheffield, Floor K, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK.
| | | | | |
Collapse
|
12
|
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
|
13
|
Schultz RM. Potential of p38 MAP kinase inhibitors in the treatment of cancer. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 60:59-92. [PMID: 12790339 DOI: 10.1007/978-3-0348-8012-1_2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The involvement of chronic inflammation in tumor development and progression is reviewed. Based on the natural history of certain diseases and epidemiology studies, a strong association has been established between particular chronic inflammatory conditions and eventual tumor appearance. Solid tumors require a stroma for their growth and recruit macrophages to synthesize essential growth and angiogenic factors that they do not have the capacity to produce. The microenvironment of the local host tissue appears to be an active participant in exchanging cytokines and enzymes with tumor cells that modify the local extracellular matrix, stimulate migration, and promote tumor angiogenesis, proliferation and survival. The role of p38 MAP kinase as a therapeutic target for treating cancer is discussed.
Collapse
Affiliation(s)
- Richard M Schultz
- Lilly Research Laboratories, Division of Cancer Research, Indianapolis, IN 46285, USA.
| |
Collapse
|
14
|
Tozer GM, Kanthou C, Parkins CS, Hill SA. The biology of the combretastatins as tumour vascular targeting agents. Int J Exp Pathol 2002; 83:21-38. [PMID: 12059907 PMCID: PMC2517662 DOI: 10.1046/j.1365-2613.2002.00211.x] [Citation(s) in RCA: 257] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The tumour vasculature is an attractive target for therapy. Combretastatin A-4 (CA-4) and A-1 (CA-1) are tubulin binding agents, structurally related to colchicine, which induce vascular-mediated tumour necrosis in animal models. CA-1 and CA-4 were isolated from the African bush willow, Combretum caffrum, and several synthetic analogues are also now available, such as the Aventis Pharma compound, AVE8062. More soluble, phosphated, forms of CA-4 (CA-4-P) and CA-1 (CA-1-P) are commonly used for in vitro and in vivo studies. These are cleaved to the natural forms by endogenous phosphatases and are taken up into cells. The lead compound, CA-4-P, is currently in clinical trial as a tumour vascular targeting agent. In animal models, CA-4-P causes a prolonged and extensive shut-down of blood flow in established tumour blood vessels, with much less effect in normal tissues. This paper reviews the current understanding of the mechanism of action of the combretastatins and their therapeutic potential.
Collapse
Affiliation(s)
- Gillian M Tozer
- Gray Cancer Institute, PO Box 100, Mount Vernon Hospital, Northwood, Middlesex HA6 2JR, UK.
| | | | | | | |
Collapse
|
15
|
Landuyt W, Ahmed B, Nuyts S, Theys J, Op de Beeck M, Rijnders A, Anné J, van Oosterom A, van den Bogaert W, Lambin P. In vivo antitumor effect of vascular targeting combined with either ionizing radiation or anti-angiogenesis treatment. Int J Radiat Oncol Biol Phys 2001; 49:443-50. [PMID: 11173139 DOI: 10.1016/s0360-3016(00)01470-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Interference with the tumor blood vessels through anti-angiogenesis or vascular targeting can indirectly suppress tumor growth. Vascular targeting of solid tumors, using tubulin-compromising agents, seems a promising and selective novel treatment. We aimed to evaluate the potential (hypothesis-based) benefit from combinations of vascular targeting using combretastatin A-4 phosphate (combreAp) with either ionizing radiation or anti-angiogenesis. METHODS AND MATERIALS Rhabdomyosarcoma tumor pieces were inplanted subcutaneously (s.c.) in the lower flank region of syngeneic adult WAG/Rij rats. Tumors were grown until different sizes and stratified for the various treatment groups: small (1-3 cm3), medium (3.1-7 cm3), and large (7.1-14 cm3). CombreAp was injected i.p.; injections of TNP-470 were s.c. in the neck area. Localized single-dose (8 Gy) irradiations of tumors were done under Nembutal anesthesia, always 1 day before a single combreAp (25 mg/kg) injection. The TNP-470 treatment (3 times 30 mg/kg in 1 week) started 1 day after a double (8 days interval between both) combreAp administration. Tumor responses were evaluated by the growth delay assay, and statistical significance of tumor growth change was computed. RESULTS Large tumors responded better to combreAp treatment given alone than did the smaller ones, confirming our previous data with this tumor model. Combining irradiation with combreAp also resulted in a tumor size-dependent growth delay. With small and medium tumor volumes, a similar response was measured after the combination treatment when compared with irradiation only. Large tumors, however, showed a strong (at least additive) increase of the growth delay with the combined therapy; the difference in tumor growth between the two treatment groups was very significant (p < 0.0001). m When TNP-470 was combined with combreAp, no significant lengthening of the growth delay, irrespective of the tumor size, was present with the applied schedule. CONCLUSION The current data show a significant advantage in the combination of combreAp with irradiation in rhabdomyosarcomas having a large size (7-14 cm3) at treatment. Such a benefit in tumor response was not observed with the smaller tumors, seemingly because irradiation as such was very effective. No significant gain in growth delay for any tumor size was observed when TNP-470, showing efficacy on its own specifically with tumors measuring <7 cm3, was added to the combreAp treatment. This presumably reflects only little angiogenesis during the first week of rhabdomyosarcoma regrowth after the combreAp treatment.
Collapse
Affiliation(s)
- W Landuyt
- Laboratory of Experimental Radiobiology, University Hospital Gasthuisberg, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hori K, Saito S, Takahashi H, Sato H, Maeda H, Sato Y. Tumor-selective blood flow decrease induced by an angiotensin converting enzyme inhibitor, temocapril hydrochloride. Jpn J Cancer Res 2000; 91:261-9. [PMID: 10761715 PMCID: PMC5926331 DOI: 10.1111/j.1349-7006.2000.tb00940.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
To enhance chemotherapeutic efficacy against cancer, it is important to deliver anticancer drugs preferentially to cancer cells and to retain the drugs there for a prolonged time. The in vivo prolongation of the exposure time of anticancer drugs in tumors can be accomplished by decreasing tumor tissue blood flow (tBF) after anticancer drug administration. The present study demonstrated that temocapril hydrochloride, an angiotensin converting enzyme inhibitor, decreases tumor tBF markedly in LY80 tumor, a subline of Yoshida sarcoma in the rat, without affecting the blood flow in liver, kidney, bone marrow, and brain. In tumor areas with flow of above 20 ml/min/100 g, the tBF decreased by approximately 50% due to temocapril. In tumor areas with tBF of about 20 ml/min/100 g, it became less than 3 ml/min/100 g with temocapril and did not recover during the 2 h experiment. These findings were obtained not only in large tumors, but also in microfoci growing within a transparent chamber. Furthermore, even when temocapril was administered under the condition of increased tumor tBF by administering angiotensin II, tumor tBF decreased immediately. Using this technique, it should be possible to trap anticancer drugs selectively in tumor tissue for an extended period of time.
Collapse
Affiliation(s)
- K Hori
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai.
| | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Hori K, Saito S, Nihei Y, Suzuki M, Sato Y. Antitumor effects due to irreversible stoppage of tumor tissue blood flow: evaluation of a novel combretastatin A-4 derivative, AC7700. Jpn J Cancer Res 1999; 90:1026-38. [PMID: 10551334 PMCID: PMC5926172 DOI: 10.1111/j.1349-7006.1999.tb00851.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The relation between tumor tissue blood flow (tBF) reduction and antitumor effects was investigated. Changes in tBF of normal tissues (liver, kidney cortex, bone marrow and brain cortex) and tumors (Yoshida sarcoma subline, LY80 and Sato lung carcinoma, SLC) due to i.v. administration of AC7700 (1, 3, 10 mg/kg), one of the combretastatin A-4 derivatives, were measured with the hydrogen clearance method. The change in blood flow in tumor microfoci was also observed directly using a rat transparent chamber. Chemotherapy against the solid tumors (LY80, SLC) was performed by administering AC7700 7 times at intervals of 3 days and the effect on the tumor growth, the histological effect, the effect on lymph node metastasis and the survival rate were investigated. Tumor tBF showed a dose-dependent response to AC7700. Although tumor tBF decreased markedly at a dose of 1 mg/kg, it tended to recover partly within several hours. At 10 mg/kg, however, tumor tBF completely stopped within approximately 30 min and never recovered in many regions. The irreversible stoppage of tumor tBF was observed in large s.c. tumors and in microfoci as well. On the other hand, in normal tissues, tBF changes due to AC7700 were not uniform. In the liver, although tBF decreased by approximately 50% at 10 mg/kg AC7700, it recovered within 8 h. In the brain, although the mean maximum reduction was 35%, the blood flow recovered to the original level within 24 h. The blood flow in the kidney cortex did not change at all. In the bone marrow, tBF decreased by approximately 80%. Generally, the blood flow reduction in normal tissues tended to be reversible. The effect on tumor growth and the histological effect were also dependent on the dose of AC7700. The tumor growth was markedly inhibited by 10 mg/ kg AC7700 and extensive necrosis was induced. Lymph node metastases were significantly inhibited and survival was prolonged significantly. In the control group, all 8 SLC tumor-bearing rats died of cancer, the presence of which was verified by gross and microscopic evaluation, within 45 days after tumor implantation. On the other hand, in the treated group, 2 of 8 rats recovered completely and survived. No obvious side effects such as body weight loss, anemia or diarrhea were observed at the dose used in this experiment. From these results, we conclude that strong antitumor effects are obtained by stopping tumor tBF irreversibly and by shutting off the nutritional supply into tumor tissue. AC7700 has been demonstrated to be a promising anticancer compound which has such an action.
Collapse
Affiliation(s)
- K Hori
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai.
| | | | | | | | | |
Collapse
|
19
|
Sersa G, Cemazar M, Parkins CS, Chaplin DJ. Tumour blood flow changes induced by application of electric pulses. Eur J Cancer 1999; 35:672-7. [PMID: 10492645 DOI: 10.1016/s0959-8049(98)00426-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effect of electric pulses on tumour blood flow was investigated in the murine fibrosarcoma SA-1. After the application of short intense electric pulses, relative tumour perfusion was measured using an 86RbC1 extraction technique. A significant reduction of tumour perfusion (approximately 30% of control) was observed within 1 h following the application of eight electric pulses to the tumour. Thereafter, tumour blood flow slowly recovered, almost reaching the pretreatment level by 24 h. No change in perfusion was induced in the untreated contralateral normal leg muscle. A similar pattern of blood flow reduction was induced when a second set of electric pulses was applied to the tumour following a 24 h interval. The degree of tumour blood flow reduction was dependent upon the number of electric pulses applied, at 1040 V, and less effect was observed if less than eight pulses were applied. A modification of the amplitude of the electric pulses resulted in changes in the direction of tumour blood flow response. Tumour blood flow increased following pulses in the range between 80 and 560 V and decreased at amplitudes higher than 640 V. These results demonstrate that the local application of electric pulses to solid tumours can modify tumour blood flow. Pulses of increased amplitude resulted in the progressive reduction of tumour blood flow with a corresponding increase in tumour cytotoxicity as measured by growth delay. Tumour blood flow reduction by electric pulses could have potential in exploiting modalities mediated by tumour hypoxia, e.g. activation of bioreductive agents.
Collapse
Affiliation(s)
- G Sersa
- Department of Tumour Biology, Institute of Oncology, Ljubljana, Slovenia.
| | | | | | | |
Collapse
|
20
|
van der Veen AH, Seynhaeve AL, Breurs J, Nooijen PT, Marquet RL, Eggermont AM. In vivo isolated kidney perfusion with tumour necrosis factor alpha (TNF-alpha) in tumour-bearing rats. Br J Cancer 1999; 79:433-9. [PMID: 10027309 PMCID: PMC2362422 DOI: 10.1038/sj.bjc.6690067] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Isolated perfusion of the extremities with high-dose tumour necrosis factor alpha (TNF-alpha) plus melphalan leads to dramatic tumour response in patients with irresectable soft tissue sarcoma or multiple melanoma in transit metastases. We developed in vivo isolated organ perfusion models to determine whether similar tumour responses in solid organ tumours can be obtained with this regimen. Here, we describe the technique of isolated kidney perfusion. We studied the feasibility of a perfusion with TNF-alpha and assessed its anti-tumour effects in tumour models differing in tumour vasculature. The maximal tolerated dose (MTD) proved to be only 1 microg TNF-alpha. Higher doses appeared to induce renal failure and a secondary cytokine release with fatal respiratory and septic shock-like symptoms. In vitro, the combination of TNF-alpha and melphalan did not result in a synergistic growth-inhibiting effect on CC 531 colon adenocarcinoma cells, whereas an additive effect was observed on osteosarcoma ROS-1 cells. In vivo isolated kidney perfusion, with TNF-alpha alone or in combination with melphalan, did not result in a significant anti-tumour response in either tumour model in a subrenal capsule assay. We conclude that, because of the susceptibility of the kidney to perfusion with TNF-alpha, the minimal threshold concentration of TNF-alpha to exert its anti-tumour effects was not reached. The applicability of TNF-alpha in isolated kidney perfusion for human tumours seems, therefore, questionable.
Collapse
Affiliation(s)
- A H van der Veen
- Department of Surgery, University Hospital Rotterdam Dijkzigt, Daniel den Hoed Cancer Centre, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
21
|
Maxwell RJ, Nielsen FU, Breidahl T, Stødkilde-Jørgensen H, Horsman MR. Effects of combretastatin on murine tumours monitored by 31P MRS, 1H MRS and 1H MRI. Int J Radiat Oncol Biol Phys 1998; 42:891-4. [PMID: 9845116 DOI: 10.1016/s0360-3016(98)00359-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PURPOSE Combretastatins have tubulin-binding activity and are being investigated for their toxicity against tumour vasculature. We report the use of 31P and 'H magnetic resonance (MR) spectroscopy and 1H MR imaging for monitoring the effects of combretastatin A-4 prodrug (100mg/kg, i.p.) on energy metabolism and necrosis, respectively, in the C3H murine mammary tumour. MATERIALS AND METHODS The tumours (volume ca. 200mm3) were grown in the hind foot of mice. MR examinations were performed without anaesthesia within a 7.1 Tesla magnet. 31P MRS (TR = 6 s) was performed before treatment and at 1-, 2-, 3-, and 24-h after injection of drug or saline via an i.p. line. 1H MRS (PRESS; 24microl voxel; TR = 2 s; TE = 135 ms) and both T1-weighted (TR = 0.2 s; TE = 0.02 s) and T2-weighted (TR = 2 s; TE = 0.20 s) 1H MRI were performed before treatment and 2.5 and 24 h afterwards. RESULTS The ratio beta-nucleotide triphosphate/inorganic phosphate fell by 33% within 1 h of treatment and remained constant for a further 2 h. A small but significant fall in pH (by 0.11 units) was observed at 1 h. Although an increase in the 1H MR spectroscopy signal at about 1.32 ppm (predominantly from lactate) was observed in some tumours following combretastatin treatment, this effect was not seen consistently. No changes in the intensity of T2-weighted 1H MR images or in tumour necrosis (measured histologically) were detected within 3 h of treatment. CONCLUSIONS The reduction in tumour energetics and pH was consistent with a reduction in tumour blood flow but this occurred before any significant incidence of haemorrhagic necrosis was detected. The combretastatin dose used to achieve these effects was less than one tenth of the maximum tolerated dose in mice.
Collapse
Affiliation(s)
- R J Maxwell
- Gray Laboratory Cancer Research Trust, Northwood, Middx, UK.
| | | | | | | | | |
Collapse
|
22
|
Chaplin DJ, Hill SA, Bell KM, Tozer GM. Modification of tumor blood flow: current status and future directions. Semin Radiat Oncol 1998; 8:151-63. [PMID: 9634492 DOI: 10.1016/s1053-4296(98)80041-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Suboptimal drug distribution and hypoxia, which can contribute to treatment failure, are a direct consequence of the spatial and temporal heterogeneity in perfusion that occurs in solid tumors. Therefore, improvements in tumor blood flow have wide-ranging therapeutic importance. Paradoxically, controlled decreases in tumor blood flow can also be exploited and, if permanent, induce extensive tumor cell death on their own. We review the current knowledge of the factors controlling tumor blood flow with emphasis on the roles of the endogeneous vasodilator nitric oxide and the endogenous vasoconstrictor endothelin-1. The potential importance and application of approaches that irreversibly damage vascular function, so-called vascular targeting, are also discussed. Emphasis is given to the drug-based approaches to vascular targeting that are now entering clinical evaluation. There is no doubt that increased understanding of the processes that determine blood flow in tumors, coupled with the availability of techniques to monitor blood flow noninvasively in the clinic, will enable strategies for selectively modifying tumor blood flow to be transferred from the laboratory to the clinical setting.
Collapse
Affiliation(s)
- D J Chaplin
- Gray Laboratory Cancer Research Trust, Mount Vernon Hospital, Northwood, Middlesex, UK
| | | | | | | |
Collapse
|
23
|
Hohenberger P, Haier J, Schlag PM. Rhabdomyolysis and renal function impairment after isolated limb perfusion--comparison between the effects of perfusion with rhTNF alpha and a 'triple-drug' regimen. Eur J Cancer 1997; 33:596-601. [PMID: 9274441 DOI: 10.1016/s0959-8049(97)00013-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The aim of this study was to monitor serum and perfusate levels of myoglobin (MB) and creatine kinase (CK) during isolated limb perfusion (ILP) in order to identify those at risk of renal failure. We investigated the release of MB and CK in 40 patients who underwent ILP for melanoma (n = 15) or sarcoma (n = 25) using rhTNF alpha/melphalan (n = 28) or a triple-drug regimen (n = 12). Serial determinations of CK and MB were performed in both perfusate and systemic circulation during and after ILP and renal function was assessed. A significant increase of MB could be detected in the perfusate during ILP. After ILP, an up to 100-fold increase with a double peak of MB at 4 h and 24 h postoperatively was observed. The maximum elevation of serum activity of CK was at 30 h. The increase for both proteins was highly significant (P < 0.001). ILP with rhTNF alpha/melphalan yielded significantly (P < 0.001) higher serum values of MB and CK and also the impairment of the renal function was more pronounced. The peak values of MB after ILP occur early and allow the patients most at risk of developing renal failure to be identified. Rhabdomyolysis can be detected early by determination of MB from the perfusate. Further measurements twice daily for 2-3 days post ILP from serum samples as well as daily assessment of MB in the urine is helpful for detecting myoglobinuria and imminent renal failure.
Collapse
Affiliation(s)
- P Hohenberger
- Division of Surgery and Surgical Oncology, Robert-Rössle Hospital and Tumor Institute, Max-Delbrück Center for Molecular Medicine, Virchow Klinikum, Humboldt University of Berlin, Germany
| | | | | |
Collapse
|
24
|
Makino M, Lodato RF, Stephens LC, Strebel FR, Jenkins G, Ohno S, Sakaguchi Y, Kostergaard J, Tomasovic SP, Bull JM. Protective effect of NG-monomethyl-L-arginine against hypotension inducted by combined tumour necrosis factor-alpha and whole body hyperthermia in rats. Int J Hyperthermia 1996; 12:617-34. [PMID: 8886889 DOI: 10.3109/02656739609027670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We studied: (a) the adverse effects of tumour necrosis factor-alpha (TNF) given during whole body hyperthermia (WBH) on mean arterial pressure (MAP) and gut mucosa in anaesthetized rats; (b) the potential protective effect of NG-monomethyl-L-arginine (L-NMA), an inhibitor of nitric oxide synthase; and (c) the influence of L-NMA on the antitumour effect of the trimodality therapy, WBH + TNF + Carboplatin (CBDCA). In normothermic rats, TNF alone (10(5) or 10(6) U/kg) did not cause hypotension, but increased MAP (p < 0.05). L-NMA alone (5, 10 and 20 mg/kg) increased MAP moderately and dose-dependently (p < 0.05). WBH (41.5 degrees C for 2 h) increased MAP markedly (from 103 +/- 4 to 161 +/- 4 mm Hg). This increase in MAP was sustained throughout the hyperthermia, but was followed by a transient relative hypotension (MAP = 80 +/- mm Hg) on cessation of WBH and an eventual return to near baseline at 30 min post-WBH (MAP = 94 +/- 5 mm Hg). WBH + TNF (10(5) or 10(6) U/kg) initially increased MAP similarly to WBH alone. During the second hour of WBH, however, MAP decreased towards pre-treatment levels, and cessation of WBH was followed by sustained hypotension. This late hypotensive state was associated with a mortality during the early (first 2 h) post-WBH period of 17 and 100% at TNF dose of 10(5) and 10(6) U/kg TNF, respectively. L-NMA given to rats receiving WBH + TNF (10(6) U/kg) maintained MAP at levels similar to WBH alone during WBH treatment. L-NMA prevented the post-WBH hypotension, and extended the survival beyond the early (first 2 h) post-WBH period. No rat, however, receiving high dose TNF (10(6) U/kg) survived more than 12 h even with L-NMA (totally 40 mg/kg). WBH + TNF (10(5) and 10(6) U/kg) also produced marked histopathological injury to the gut mucosa at 2 h post-treatment. L-NMA substantially protected the gut from this injury. In rats bearing a transplantable fibrosarcoma, L-NMA did not decrease the antitumour effect consisting of WBH + TNF (10(5) U/kg) + CBDCA, while it decreased (p < 0.05) the general toxicity (weight loss, diarrhea and foot oedema) of this combination. We conclude that L-NMA may prevent or ameliorate the early toxicity but not the late lethal effects of WBH + high dose TNF (10(6) U/kg). Additionally, L-NMA reduces some of the toxicity of WBH + TNF (10(5) U/kg) + CBDCA without decreasing the antitumour effect of this trimodality therapy. Inhibitors of nitric oxide synthase such as L-NMA may provide a novel approach to overcoming the toxicity of TNF in combination with WBH.
Collapse
Affiliation(s)
- M Makino
- Department of Internal Medicine, University of Texas Medical School, Houston, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chaplin DJ, Pettit GR, Parkins CS, Hill SA. Antivascular approaches to solid tumour therapy: evaluation of tubulin binding agents. THE BRITISH JOURNAL OF CANCER. SUPPLEMENT 1996; 27:S86-S88. [PMID: 8763854 PMCID: PMC2150027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
We have assessed the vascular effects of vinblastine and four other tubulin binding agents (dolastatin 10, dolastatin 15, combretastatin A1 and combretastatin A4), which are awaiting clinical evaluation. All five agents induce a reduction in tumour blood flow as measured by uptake of RbCI 24 h post drug administration. The degree of reduction ranged from 50% with combretastatin A1 to 90% with dolastatin 10. These reductions were similar to that seen with flavone acetic acid (FAA) and indicate that antivascular effects are a common feature of tubulin binding agents. We subsequently evaluated whether the blood flow reductions, induced by FAA and vinblastine, could be used to enhance the activity of the bioreductive drug tirapazamine. Since the kinetics and extent of blood flow reductions induced by the agents is comparable, similar therapeutic response was expected. Potentiation was only evident with FAA, indicating that this effect is not directly related to killing of hypoxic tumour cells induced as a consequence of blood flow reduction.
Collapse
Affiliation(s)
- D J Chaplin
- Tumour Microcirculation Group, Gray Laboratory Cancer Research Trust, Mount Vernon Hospital, Northwood, Middlesex
| | | | | | | |
Collapse
|
26
|
Lin JC, Park HJ, Song CW. Combined treatment of IL-1 alpha and TNF-alpha potentiates the antitumour effect of hyperthermia. Int J Hyperthermia 1996; 12:335-44. [PMID: 9044903 DOI: 10.3109/02656739609022522] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vasoactive cytokines, such as IL-1 alpha and TNF-alpha, modulate the homeostatic state at the endothelial surface and cause various types of pathological damage in vascular systems. We investigated the potential therapeutic effects of IL-1 alpha and TNF-alpha in combination with hyperthermia on SCK tumours grown in the legs of A/J mice. We first determined the effect of cytokines on tumour blood perfusion with the (86)Rb uptake method. When the host mice were given an i.p. injection of 25 mu g/kg IL-1 alpha or 50 mu g/kg TNF-alpha, the tumour blood perfusion markedly declined to 46 and 82% of control, respectively. The combination of IL-1 alpha and TNF-alpha reduced the 86Rb uptake to 41% bf control. Hyperthermia at 42.5 degrees C for 1 h reduced the tumour blood flow to 71% of control. The tumour blood perfusion decreased further to 20% of control when the tumours were heated for 1 h at 42.5 degrees C starting 4h after the injection of both IL-1 alpha and TNF-alpha. The changes in clonogenic cell numbers in SCK tumours, as determined by the in vivo-in vitro assay, following various treatments was also investigated. At 4 h after an i.p. injection of 25 mu g/kg IL-1 alpha or 50 mu g/kg TNF-alpha, the clonogenicity of SCK tumours significantly decreased to 29 or 37% of control, respectively. Heating at 42.5 degrees C for 1 h caused a decline in the clonogenic cell number to 30% of control. When both IL-1 alpha and TNF-alpha were given and tumours were heated 4h later at 42.5 degrees C for 1 h, the clonogenic cell number markedly declined to 0.4% of control. The time needed for control tumours to reach 4 x their initial volume was about 3 days, and treatment with IL-1 alpha or hyperthermia alone induced a tumour delay growth by about 1 day. The combined injection of IL-1 alpha and TNF-alpha followed by a heating at 42.5 degrees C for 1 h delayed the tumour growth by 6 days. The results in this study suggest that prior impairment of blood circulation by the combined treatment of IL-1 alpha and TNF-alpha potentiates hyperthermic damage in tumours.
Collapse
Affiliation(s)
- J C Lin
- University of Minnesota Medical School, Department of Therapeutic Radiology-Radiation Oncology, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
27
|
Abstract
The tumour vasculature is vital for the establishment, growth and metastasis of solid tumours. Its physiological properties limit the effectiveness of conventional anti-cancer strategies. Therapeutic approaches directed at the tumour vasculature are reviewed, suggesting the potential of anti-angiogenesis and the targeting of vascular proliferation antigens as cancer treatments.
Collapse
Affiliation(s)
- C T Baillie
- University Department of Surgery, Royal Free Hospital and School of Medicine, London, UK
| | | | | |
Collapse
|
28
|
Dahn MS, Lange MP, Kosir MA. Splanchnic metabolism associated with liver metastasis. Intensive Care Med 1995; 21:352-5. [PMID: 7650259 DOI: 10.1007/bf01705415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Metastatic liver disease can modify the metabolic response to critical illness. Systemic lactic acidosis may arise from an increased production due to inadequate peripheral tissue oxygen transport, altered metabolic function such as depressed pyruvate oxidation or insufficient hepatic clearing capacity due to tumor replacement of functional liver mass. Hepatic venous catheterization in a patient with extensive metastatic melanoma to the liver and adult respiratory distress syndrome indicated a marked disparity between whole body and liver oxygenation which may arise due to a markedly stepped up splanchnic oxygen utilization unmatched by a proportionate rise in regional oxygen delivery. Since some neoplasms may exhibit increased metabolic activity, it is suspected that these metastatic lesions may have contributed to the observed regional hypermetabolism thereby worsening hepatic hypoxia and exacerbating lactic acidosis. This case also illustrates the difficulties in interpreting global indicators of metabolic function and oxygenation in critically ill patients.
Collapse
Affiliation(s)
- M S Dahn
- Department of Surgery, University Health Center, Detroit, MI 48201, USA
| | | | | |
Collapse
|
29
|
Rowlinson-Busza G, Maraveyas A, Epenetos AA. Effect of tumour necrosis factor on the uptake of specific and control monoclonal antibodies in a human tumour xenograft model. Br J Cancer 1995; 71:660-5. [PMID: 7710925 PMCID: PMC2033735 DOI: 10.1038/bjc.1995.131] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The investigations reported in this paper aim to exploit tumour necrosis factor (TNF)-induced vascular changes in an attempt to increase the tumour uptake of specific monoclonal antibody. The vascular permeability to monoclonal antibody of a human tumour xenograft increased 2.6-fold by 1 h post injection of 2.5 x 10(3) U of TNF, although this effect was lost by 3 h. The normal tissues also demonstrated increased vascular permeability to IgG, but to a lesser extent. Liver permeability increased 1.5-fold at 1 h but returned to the control value by 6 h. Lung permeability increased 1.4-fold at 1 h post injection and returned to normal by 3 h. Muscle values were not significantly increased compared with controls. The blood activity was cleared more quickly in the TNF-treated mice (t1/2 beta = 101 h, compared with 121 h in control mice). This was probably due to the increased vascular permeability in normal organs of treated mice. At 1 day and 3 days post injection, the tumour uptake of the specific, but not the control, antibody was significantly increased by 25% and 29% respectively. This resulted in an increase in the area under the tumour activity curve, and therefore tumour radiation dose, of 25% in treated compared with control mice. In addition, a consequence of the faster blood clearance of the isotope in the TNF-treated mice was a reduction in the area under the blood activity curve of 12%, thereby reducing systemic toxicity. The increase in vascular permeability to IgG following TNF injection resulted in both specific and control antibodies having improved access to the tumour antigens, and a transient increase in uptake was observed. Only in the case of the specific antibody was the increase maintained, since this antibody binds to the available antigenic sites, whereas the control antibody was cleared from the tumour without binding. No evidence of tumour necrosis was observed at the TNF doses given, nor was there any toxicity to the mice.
Collapse
Affiliation(s)
- G Rowlinson-Busza
- Imperial Cancer Research Fund Oncology Unit, Royal Postgraduate Medical School, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
30
|
Dahl O. Interaction of Heat and Drugs In Vitro and In Vivo. THERMORADIOTHERAPY AND THERMOCHEMOTHERAPY 1995. [DOI: 10.1007/978-3-642-57858-8_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
31
|
Sasajima T, Mineura K, Sasaki J, Kowada M, Tomura N, Hatazawa J, Ogawa T, Uemura K. Positron emission tomographic assessment of cerebral hemocirculation and glucose metabolism in malignant glioma following treatment with intracarotid recombinant human tumor necrosis factor-alpha. J Neurooncol 1995; 23:67-73. [PMID: 7623071 DOI: 10.1007/bf01058461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cerebral hemocirculation and glucose metabolism in a malignant astrocytoma were repeatedly quantified before and after intracarotid injection of recombinant human tumor necrosis factor-alpha (rH-TNF) using positron emission tomography (PET). The patient received an intracarotid injection of a 3 x 10(4) U/m2 dose of rH-TNF three times over a two week period. PET was performed prior to and 24 hr after the first injection, and two weeks after the third injection. Prior to the first rH-TNF treatment, two lesions demonstrating high perfusion and hypermetabolism of glucose were noted in the right frontal and temporal regions. The frontal hypermetabolic lesion showed decreases in hemocirculation and metabolism 24 hr after the first injection and then increases beyond the pre-treatment level two weeks after the third treatment, whereas the temporal lesion remained unchanged during the follow-up period. No appreciable changes were noted in the adjacent cortex where rH-TNF was perfused, with the exception of a transient decrease in regional blood volume. Magnetic resonance images of the tumor showed no changes as a result of treatment with intracarotid rH-TNF. Intracarotid rH-TNF preferentially affects tumor tissue as opposed to normal cortex.
Collapse
Affiliation(s)
- T Sasajima
- Neurosurgical Service, Akita University Hospital, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Engel T, Vaupel P. Acute effects of tumor necrosis factor-alpha or lymphotoxin on oxygenation and bioenergetic status in experimental tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1994; 345:501-8. [PMID: 8079750 DOI: 10.1007/978-1-4615-2468-7_67] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- T Engel
- Institute of Physiology and Pathophysiology, University of Mainz, Germany
| | | |
Collapse
|
33
|
Chaplin DJ, Horsman MR. The influence of tumour temperature on ischemia-induced cell death: potential implications for the evaluation of vascular mediated therapies. Radiother Oncol 1994; 30:59-65. [PMID: 8153381 DOI: 10.1016/0167-8140(94)90010-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have evaluated the influence of tumour temperature on the kinetics and extent of tumour cell death following induction of ischemia. Induction of ischemia in SCCVII tumours implanted subcutaneously in the back of syngeneic C3H mice results in a rapid cooling of the tumour from a resting value of 35.2 degrees C, towards ambient temperature. In the SCCVII tumour no significant cell kill is detected in the first hour of ischemia. At longer times cell kill was detected and a survival level of 2 x 10(-2) was attained after 6 h of ischemia. However, if the tumours were maintained at a temperature of 37 degrees C following induction of ischemia a more rapid and dramatic reduction in cell survival is observed with a survival level of approximately 10(-5) being attained after 4 h of ischemia. Qualitatively similar results are obtained using the Lewis lung tumour implanted in C57BL mice. Similar rapid cooling following occlusion of the blood supply is observed in the C3H/TIF tumour implanted in the foot. For the C3H/TIF induction of ischemia for up to 6 h resulted in no significant growth delay provided the tumours were kept at room temperature. However, if the tumours were maintained at a temperature of 37 degrees C during the ischemic insult significant growth delay was observed for all clamping times exceeding 1 h. These results show the importance of tumour temperature on the kinetics and extent of tumour cell kill during ischemia. This finding has particular relevance for studies in which agents known to reduce tumour blood flow are used in animals bearing superficially located tumours.
Collapse
Affiliation(s)
- D J Chaplin
- CRC Gray Laboratory, Mount Vernon Hospital, Northwood Middlesex, UK
| | | |
Collapse
|
34
|
Naredi PL, Lindnér PG, Holmberg SB, Stenram U, Peterson A, Hafström LR. The effects of tumour necrosis factor alpha on the vascular bed and blood flow in an experimental rat hepatoma. Int J Cancer 1993; 54:645-9. [PMID: 8514456 DOI: 10.1002/ijc.2910540420] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The influence of TNF alpha on tumour growth rate has been attributed to its effects on the vascular bed and blood flow. The aim of our study was to investigate the effects of pharmacological doses of TNF alpha on the tumour vascular bed and to quantify blood flow in an experimental hepatoma during a more extended period after TNF-alpha exposure than hitherto reported. In Lister rats, a syngeneic rat hepatoma was implanted on the dorsum of the right hind foot. TNF alpha was given i.v. The injection was repeated after 24 hr. Tumour blood flow was estimated before and 1, 24, and 96 hr after TNF-alpha administration with the 133Xe-washout technique. The passage of microspheres through the tumour vascular bed (non-entrapment), as a measure of vascular occlusion, was estimated 4 and 96 hr after TNF-alpha administration. Tumour growth rate was measured. The tumours were subjected to histological examination and the sensitivity to TNF alpha in vitro was tested. A reduction of tumour blood flow was observed in TNF-alpha-treated groups. Tumour growth rate was equally increased after 96 hr in both the TNF-alpha groups as compared with controls. There was no significant change in non-entrapment for the TNF-alpha-treated rats as compared with controls. Histology revealed extensive necrosis and thrombosis in tumours. TNF alpha had no effect on the viability of the cloned hepatoma cell line in vitro.
Collapse
Affiliation(s)
- P L Naredi
- Department of Surgery, Sahlgrenska Hospital, University of Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
35
|
Folli S, Pèlegrin A, Chalandon Y, Yao X, Buchegger F, Lienard D, Lejeune F, Mach JP. Tumor-necrosis factor can enhance radio-antibody uptake in human colon carcinoma xenografts by increasing vascular permeability. Int J Cancer 1993; 53:829-36. [PMID: 8449608 DOI: 10.1002/ijc.2910530521] [Citation(s) in RCA: 84] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Marked differences in the tumor uptake of a 125I-labeled monoclonal antibody (MAb) directed against carcinoembryonic antigen (CEA) were observed in 4 serially transplanted human colorectal carcinomas in nude mice. A comparative study showed that elevated values of measurable tumor vascular parameters, such as permeability, blood flow and blood volume, correlated better with high MAb tumor uptake than the concentration of target antigen in the tumor. In an attempt to modify the vascular parameters and to determine if this could increase antibody uptake by the tumor, rhTNF alpha (TNF) was injected i.t. or i.v. and antibody localization experiments were performed immediately thereafter. Results showed that the permeability of the tumor vessels increased 8 to 10 fold 1 hr after i.t. injection of TNF as compared to control tumors injected with saline. Tumor uptake of 125I-labeled anti-CEA MAb, was 3 times higher 2 hr after i.v. injection and still 27% higher 22 hr later, as compared to results from controls. Intravenous injection of TNF simultaneously with the 125I-labeled anti-CEA MAb also resulted in a 2-fold increase in tumor uptake 4 hr after injection, but the increase was no longer significant 24 hr after injection. Interestingly after i.v. injection of TNF, the MAb concentration in the blood and other normal tissues, such as liver, kidneys, lungs and heart was decreased, resulting in significantly higher ratios of tumor to normal tissue. Taken together the results demonstrate that injection of TNF can increase tumor vascular permeability and improve radio-antibody uptake. This raises the possibility of increasing the radiation dose delivered by antibody to the tumor in the course of radioimmunotherapy.
Collapse
Affiliation(s)
- S Folli
- Institute of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Svitkina TM, Parsons DF. Binding of some metastatic tumor cell lines to fibrous elastin and elastin peptides. Int J Cancer 1993; 53:824-8. [PMID: 8383630 DOI: 10.1002/ijc.2910530520] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recent suggestions that tumor-cell targeting of elastin-rich tissues (e.g., lung) correlates with the presence of surface elastin receptors have been investigated. Receptors for insoluble (fibrous) elastin and for soluble elastin peptides have been implemented in these correlations. A rapid assay for binding of insoluble elastin has been devised. Two of the cell lines tested (M27 and MAT-LyLu), which metastasize to the lung, strongly bound fibrous elastin whereas a third (B16-F10) did not. None of 4 metastatic cell lines that do not target the lung (A549, 3LL, TA3, TA3-iso2) bound fibrous elastin. The ability of cell lines to interact with soluble elastin was tested by cell attachment to high-molecular-weight soluble elastin peptides adsorbed on a plastic surface. Three of 7 tested cell lines, B16-F10, M27 and TA3, attached to a soluble elastin coating. In contrast to the rapid binding of insoluble elastin particles, the cell interaction with immobilized soluble elastin peptides was delayed, suggesting that induction of receptors for soluble elastin and/or modification of the elastin coat was occurring. Thus, all 3 tested cell lines where metastases target the lung, namely, MAT-LyLu, B16-F10 and M-27, show soluble- or insoluble-elastin interactions, whereas, of 4 cell lines not targeting lungs, only one, TA3, reacts with soluble elastin.
Collapse
Affiliation(s)
- T M Svitkina
- A.N. Belozersky Institute of Physical and Chemical Biology, Moscow State University, Russia
| | | |
Collapse
|
37
|
Denekamp J. Review article: angiogenesis, neovascular proliferation and vascular pathophysiology as targets for cancer therapy. Br J Radiol 1993; 66:181-96. [PMID: 7682469 DOI: 10.1259/0007-1285-66-783-181] [Citation(s) in RCA: 242] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A body of evidence that vascular-mediated damage occurs in murine tumours after many existing forms of anti-tumour therapy is rapidly accumulating (see Gray Conference Proceedings edited by Moore & West, 1991). Rapid conventional screens of cells in vitro or using leukaemias of lymphomas will not detect this mode of action and such screens will therefore miss effective agents. A change in the approach to experimental cancer therapy is needed to ensure that this important new avenue is fully investigated. Solid tumours will need to be studied and the importance of specific tumour cell biochemistry (e.g. on tissue factor procoagulant activity), of endothelial status and the immunocompetence of the host are all likely to be important. It is a subject of considerable debate at present whether transplanted subcutaneous mouse tumours are adequate models and whether they will reflect the response of spontaneous tumours, or even of transplants into other sites. Xenografts are not likely to be appropriate if the immuno-suppressed hosts lack the cells needed for the cytokine component of the pathway. The strategy of design and screening of new agents, for scheduling of existing agents and particularly the sequencing of adjunctive therapies are likely to be completely different for the "direct" tumor cell or "indirect" vascular-mediated approaches. It may eventually be appropriate to combine vascular manipulation with direct cytotoxicity aimed at malignant cells but the two mechanisms must be recognized as distinct entities and considered separately before attempting to coordinate them. It is important therefore to identify the "hallmarks" of vascular mediated injury and the means by which this can be distinguished from direct cell kill. These may be detectable in the tumour response but clues can also be gained from the side effects that are seen in normal tissues both with existing and with novel therapies (Figure 7). The appeal of vascular-mediated ischaemic therapy is that it is systemic and will have the potential of being effective on any tumour with a newly evoked vascular network, i.e. of about 1 mm in diameter, but it will be even more effective on large tumours than on small. Thus it should affect both large primary tumours and disseminated small metastases. The studies with many different anti-cancer agents have illustrated the potential complexity of responses that can appear to cause tumour cell death by collapse or occlusion of the blood supply. They have also focused attention on features of disparate agents, e.g. TNF, FAA, PDT, which may share similar pathways. No single feature of neovasculature can be highlighted as the sole route by which such antivascular therapy should be targeted. Rapid proliferation of the endothelial cells may prove to be a target, but it also influences differentiation characteristics, so that the immature cells will function abnormally. The permeability of these poorly formed vessels may lead to extravasation of proteins leading to increase interstitial pressures and by this means to an imbalance between intravascular and extravascular pressures and hence to collapse of the thin-walled vessels. Changes in systemic blood pressure, cardiac output, viscosity or coagulation and especially a redistribution of regional perfusion would all have differential effects in tumours and normal vessels. Clearly both vascular patho-physiology and the complexity of endothelial cell function and its imbalance in neovasculature will be important in understanding the mechanism of action of antivascular strategies. This very challenging boundary between oncology and a number of other medical and biological fields promises to lead to altered attitudes to existing therapies and the discovery of completely new classes of anti-cancer agents. The next decade should translate into clinical benefit for patients if the progress in this field continues to be as rapid as it has been in the late eighties. We must now determine what characteristics make one tumour more sensitive than another to agents such as heat, PDT, cytokines and FAA, and learn how to extrapolate from those rodent tumours to the human.
Collapse
Affiliation(s)
- J Denekamp
- CRC Gray Laboratory, Mount Vernon Hospital, Northwood, Middlesex, UK
| |
Collapse
|
38
|
Affiliation(s)
- C S Johnson
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pennsylvania
| |
Collapse
|
39
|
|
40
|
Abstract
Tumor necrosis factor (TNF) is a multifunctional cytokine which has excited and fascinated numerous investigators and commercial entities due to its promise as a therapeutic agent against cancer and as a target for drugs treating septic shock. TNF is a protein having cytotoxic, cytostatic, immunomodulatory as well as several other activities and is also involved in septic shock. This review covers the structure of TNF and its receptors, various in vitro activities and in vivo activities based on studies in animal model systems. The role of TNF as an anticancer therapeutic agent, based on various phase I and phase II clinical studies, has also been considered. The review concludes with several considerations for increasing the therapeutic utility of TNF in terms of targeting, toxicity and half-life.
Collapse
Affiliation(s)
- R S Sidhu
- Cytoclonal Pharmaceutics Inc., Dallas, TX 75235
| | | |
Collapse
|
41
|
Pimm MV, Gribben SJ, Morris TM. Influence of recombinant tumour necrosis factor alpha on blood flow and antibody localisation in human tumour xenografts in nude mice. J Cancer Res Clin Oncol 1991; 117:543-8. [PMID: 1744160 DOI: 10.1007/bf01613286] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mice with s.c. grafts of gastric carcinoma MKN45 or osteosarcoma 788T were injected i.v. with recombinant tumour necrosis factor alpha (rTNF alpha) and tumour blood flow rates were determined 4 h later as a fraction of the cardiac output g tissue-1. With MKN45, the tumour blood flow rate was significantly reduced from a mean of 1.86% cardiac output g-1 to 0.84% and 0.65% with 50 and 200 micrograms kg-1 rTNF alpha respectively. With 788T, the tumour blood flow rate was reduced at 50 micrograms kg-1 rTNF alpha from 1.13% cardiac output g-1 to 0.56%. There were essentially no changes in blood flow rates in other organs. The effect of rTNF alpha on localisation of monoclonal antibodies into these xenografts were examined. When a single dose of rTNF alpha (50 micrograms kg-1) was given at the same time as labelled NCRC-2 antibody there was a significant reduction in localisation into 788T osteosarcoma xenografts. In other tests, mice were injected daily for 3 days with 50 micrograms kg-1 rTNF alpha. They were injected i.v. with monoclonal antibody 4 h after the first injection and dissected on the 4th day. With 788T there was a small but not statistically significant reduction in the absolute amount of NCRC-2 antibody localising in tumour, although this reduction was greater when results were expressed as tumour-to-blood ratios. With MKN45 xenografts, treatment with rTNF alpha had little effect on tumour localisation of an anti-(carcinoembryonic antigen) monoclonal antibody (NCRC-24). These studies show that TNF can be administered so as to reduce tumour blood flow and with little effect on tumour localisation of antibody, suggesting that combination therapy with TNF and antibodies or their immunoconjugates is feasible. Other studies have suggested that TNF can increase antibody localisation into tumours, but this was not seen here, and in some cases administration could reduce tumour localisation. It appears that this method of enhancing antibody localisation may be critically dependent on scheduling, and therefore it may not be extensively applicable.
Collapse
Affiliation(s)
- M V Pimm
- Cancer Research Campaign Laboratories, University of Nottingham, UK
| | | | | |
Collapse
|
42
|
Kiwit JC, Schmitz KH, Daum L, Reifenberger G, Roosen N. Effects of recombinant human tumor necrosis factor on rodent gliomas and normal brain. J Neurosurg 1991; 75:597-603. [PMID: 1653310 DOI: 10.3171/jns.1991.75.4.0597] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In a study examining the possible therapeutic effects of recombinant human tumor necrosis factor-alpha (rHuTNF-alpha) on malignant gliomas without expression of tumor necrosis factor (TNF)-receptors, RG-2 glioma cells were tested in vitro as well as in a rat experimental glioma model. A growth inhibition assay revealed no inhibiting effect in vitro up to a concentration of 20 micrograms/ml rHuTNF-alpha. Receptor-binding studies showed that RG-2 cells did not present specific receptors for rHuTNF-alpha. The pharmacokinetics of rHuTNF-alpha after intravenous injection were studied with respect to serum, tissue, and brain tumor concentrations and showed increased glioma concentrations of (mean +/- standard error of the mean) 0.47 +/- 0.18 ng TNF/mg brain compared to 0.15 +/- 0.05 ng TNF/mg brain in the normal contralateral hemisphere. No therapeutic effect on solid RG-2 gliomas could be observed after stereotactic injection of 7.3 micrograms rHuTNF/10 microliter buffer solution into the tumor in 10 animals. Immunohistochemical studies after stereotactic injection of rHuTNF-alpha showed total disappearance of the substance after 24 hours without internalization into tumor cells. Stereotactic injection of 7.3 micrograms rHuTNF 10 microliters into normal brain resulted in marked inflammatory response around the injection track, including microvascular thrombosis. These results demonstrate that rHuTNF has neither direct nor indirect cytotoxic activity on RG-2 glioma cells. Furthermore, before clinical use of rHuTNF-alpha in malignant gliomas, the authors suggest that receptor studies be done in each patient. In receptor-positive patients undergoing treatment with rHuTNF-alpha, precautions should be taken to prevent local encephalitic reactions.
Collapse
Affiliation(s)
- J C Kiwit
- Neurosurgical Clinic, Heinrich-Heine-University Medical School, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
43
|
Abstract
It has been established that malignant tissue as a consequence of abnormal morphogenesis has a structurally abnormal blood supply. These structural and as a consequence functional differences between normal and neoplastic vasculature provide a basis for selective modulation of tumour vascular function. Agents have been identified which can induce both irreversible and reversible effects on tumour blood flow. Hyperthermia, photodynamic therapy, tumour necrosis factor and flavone acetic acid are known to elicit most of their anti-tumour effect via irreversible changes in tumour vascular function. In addition to the extensive tumour cell kill and thus therapeutic potential provided by such chronic modulation of blood flow, acute transient changes in macroregional and microregional tumour blood flow could also play an important role if used appropriately with conventional therapies. The use of this latter type of modulation is discussed with reference to known examples of such 'vasoactive' compounds. It is also emphasized that blood flow changes induced in tumour tissue can be a 'double-edged sword' with detrimental consequences for therapeutic outcome if inappropriate changes are induced, for example, reductions in flow at the time of conventional radiotherapy or chemotherapy by agents not considered to be 'vasoactive'. To emphasize this point examples of blood flow modulation by pimonidazole and cis-platinum, agents that are used in conjunction with radiotherapy, are described.
Collapse
Affiliation(s)
- D J Chaplin
- Medical Biophysics Unit, B.C. Cancer Research Centre, Vancouver, Canada
| |
Collapse
|
44
|
Edwards HS, Bremner JC, Stratford IJ. Induction of hypoxia in the KHT sarcoma by tumour necrosis factor and flavone acetic acid. Int J Radiat Biol 1991; 59:419-32. [PMID: 1671692 DOI: 10.1080/09553009114550381] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The interaction of FAA or TNF with radiation was studied in the murine KHT sarcoma. When used alone both agents showed a dose- and time-dependent toxicity towards the tumour cells and significantly reduced tumour blood flow within 1 h of treatment. When used in combination with radiation, both TNF and FAA caused an increase in the fraction of hypoxic cells in the KHT tumour. This was assessed by an in vivo/in vitro clonogenic assay and by a comparison with the radioprotection provided by clamping tumours prior to and during irradiation. When TNF was given at a dose of 2.5 X 10(5) U/kg an increase in tumour hypoxia was seen after 30 min. Close to 100% radiobiological hypoxia was reached by 1 h after treatment, lasting for up to 16 h. Doses of TNF below 0.25 X 10(5) U/kg did not induce levels of hypoxia comparable to clamping when administered 3 h prior to irradiation. Similarly, FAA produced a rapid increase in tumour hypoxia: a dose of 200 mg/kg induced close to 100% radiobiological hypoxia when give 1 h prior to irradiation. Complete tumour hypoxia was still apparent 18 h after treatment with FAA. Administered doses of FAA below 100 mg/kg did not produce close to 100% radiobiological hypoxia when administered 3 h prior to irradiation.
Collapse
Affiliation(s)
- H S Edwards
- Medical Research Council Radiobiology Unit, Chilton, Didcot, Oxford, UK
| | | | | |
Collapse
|
45
|
Affiliation(s)
- J Denekamp
- CRC Gray Laboratory, Mount Vernon Hospital, Northwood, Middlesex, U.K
| | | |
Collapse
|
46
|
Abstract
The blood supply to all solid tumours consists of parasitized normal vessels and new vessels which have been induced to grow by the presence of the tumour. These vessels are inadequate in many respects, being tortuous, thin-walled, chaotically arranged, lacking innervation and with no predetermined direction of flow. The walls consist of a basement membrane lined with rapidly proliferating immature endothelial cells, and are more permeable than normal vessels. The spacing of the vessels and their average diameters are not optimal for nutrient provision. This paper focuses on the evidence that many existing therapies may already have, as part of their action, a vascular mediated process of killing tumour cells. This may result from local changes within individual vessels or from systemic alterations in blood pressure, viscosity, coagulability etc. The hallmarks of vascular injury are identified and the dangers of discarding useful anticancer agent by failing to understand their mechanism of action are highlighted.
Collapse
Affiliation(s)
- J Denekamp
- CRC Gray Laboratory, Mount Vernon Hospital, Northwood, Middlesex, UK
| |
Collapse
|