The hypoxic cell: a target for selective cancer therapy--eighteenth Bruce F. Cain Memorial Award lecture.

It has been appreciated for more than 50 years that very low levels of oxygenation, or hypoxia, both protect cells from killing by X-irradiation and are present in solid tumors but not in normal tissues. Until recently, however, there has been no definitive proof that hypoxia in human tumors contributes to radiotherapy treatment failure. We now know that hypoxia in solid tumors is not only a major problem for radiation therapy but also leads to resistance to most anticancer drugs and, importantly, appears to accelerate malignant progression and increase metastasis. To date, efforts to overcome the problem of hypoxia have had only limited success. However, the recent development of new drugs that are nontoxic until they are activated in the hypoxic cell opens a new era. The first of these new drugs to be tested clinically, tirapazamine, a drug that is highly toxic to hypoxic but not aerobic cells, has already demonstrated efficacy in selective potentiation of cisplatin in randomized Phase III trials with non-small cell lung cancer. The unique presence of hypoxic cells in human tumors provides an important target for selective cancer therapy.

[1]  P. Cook The organization of replication and transcription. , 1999, Science.

[2]  B. Wouters,et al.  Cisplatin anti-tumour potentiation by tirapazamine results from a hypoxia-dependent cellular sensitization to cisplatin , 1999, British Journal of Cancer.

[3]  D A Bloch,et al.  Tissue oxygen distribution in head and neck cancer patients , 1999, Head & neck.

[4]  S. Spencer,et al.  Concurrent tirapazamine and radiotherapy for advanced head and neck carcinomas: a Phase II study. , 1998, International journal of radiation oncology, biology, physics.

[5]  C. Langer,et al.  Tirapazamine with cisplatin in patients with advanced non-small-cell lung cancer: a phase II study. , 1998, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[6]  G. Semenza Hypoxia-inducible factor 1: master regulator of O2 homeostasis. , 1998, Current opinion in genetics & development.

[7]  M C Weissler,et al.  Hypoxia and vascular endothelial growth factor expression in human squamous cell carcinomas using pimonidazole as a hypoxia marker. , 1998, Cancer research.

[8]  H. Lyng,et al.  Tumour hypoxia and vascular density as predictors of metastasis in squamous cell carcinoma of the uterine cervix. , 1998, British Journal of Cancer.

[9]  A. Fyles,et al.  Oxygenation predicts radiation response and survival in patients with cervix cancer. , 1998, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[10]  D. Chaplin,et al.  Microenvironmental control of gene expression: implications for tumor angiogenesis, progression, and metastasis. , 1998, Seminars in radiation oncology.

[11]  P. Glazer,et al.  Mutagenesis induced by the tumor microenvironment. , 1998, Mutation research.

[12]  J. Brown,et al.  Tirapazamine is metabolized to its DNA-damaging radical by intranuclear enzymes. , 1998, Cancer research.

[13]  M. Kris,et al.  Phase II study of the combination of the novel bioreductive agent, tirapazamine, with cisplatin in patients with advanced non-small-cell lung cancer. , 1997, Annals of oncology : official journal of the European Society for Medical Oncology.

[14]  A. Harris,et al.  Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[15]  M. Dewhirst,et al.  Tumor hypoxia adversely affects the prognosis of carcinoma of the head and neck. , 1997, International journal of radiation oncology, biology, physics.

[16]  B. Wouters,et al.  Cells at intermediate oxygen levels can be more important than the "hypoxic fraction" in determining tumor response to fractionated radiotherapy. , 1997, Radiation research.

[17]  P. Glazer,et al.  Genetic instability induced by the tumor microenvironment. , 1996, Cancer research.

[18]  P Vaupel,et al.  Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. , 1996, Cancer research.

[19]  P. Hoskin,et al.  Microregional blood flow in murine and human tumours assessed using laser Doppler microprobes. , 1996, The British journal of cancer. Supplement.

[20]  M. Dewhirst,et al.  Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. , 1996, Cancer research.

[21]  David E. Housman,et al.  Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours , 1996, Nature.

[22]  C. Koch,et al.  Identification of hypoxia in cells and tissues of epigastric 9L rat glioma using EF5 [2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl) acetamide]. , 1995, British Journal of Cancer.

[23]  W. Curran,et al.  Results of an RTOG phase III trial (RTOG 85-27) comparing radiotherapy plus etanidazole with radiotherapy alone for locally advanced head and neck carcinomas. , 1995, International journal of radiation oncology, biology, physics.

[24]  R. Chin,et al.  Metallothionein IIA is up-regulated by hypoxia in human A431 squamous carcinoma cells. , 1994, Cancer research.

[25]  Durand Re The influence of microenvironmental factors during cancer therapy. , 1994 .

[26]  G L Rosner,et al.  Pretreatment oxygenation profiles of human soft tissue sarcomas. , 1994, International journal of radiation oncology, biology, physics.

[27]  T. Graeber,et al.  Hypoxia induces accumulation of p53 protein, but activation of a G1-phase checkpoint by low-oxygen conditions is independent of p53 status , 1994, Molecular and cellular biology.

[28]  W. Denny,et al.  Exploiting tumor hypoxia through bioreductive release of diffusible cytotoxins: the cobalt(III)-nitrogen mustard complex SN 24771. , 1994, International journal of radiation oncology, biology, physics.

[29]  M. Dorie,et al.  Tumor-specific, schedule-dependent interaction between tirapazamine (SR 4233) and cisplatin. , 1993, Cancer research.

[30]  J. Brown,et al.  SR 4233 (tirapazamine): a new anticancer drug exploiting hypoxia in solid tumours. , 1993, British Journal of Cancer.

[31]  E. Keshet,et al.  Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis , 1992, Nature.

[32]  J. Brown,et al.  Repair of DNA and chromosome breaks in cells exposed to SR 4233 under hypoxia or to ionizing radiation. , 1992, Cancer research.

[33]  B. Price,et al.  Gadd45 and Gadd153 messenger RNA levels are increased during hypoxia and after exposure of cells to agents which elevate the levels of the glucose-regulated proteins. , 1992, Cancer research.

[34]  P. Vaupel,et al.  Oxygenation of carcinomas of the uterine cervix: evaluation by computerized O2 tension measurements. , 1991, Cancer research.

[35]  R. Mason,et al.  Microsomal reduction of 3-amino-1,2,4-benzotriazine 1,4-dioxide to a free radical. , 1991, Molecular pharmacology.

[36]  P Vaupel,et al.  Oxygenation of human tumors: evaluation of tissue oxygen distribution in breast cancers by computerized O2 tension measurements. , 1991, Cancer research.

[37]  E. Pettersen,et al.  Cell inactivation and cell cycle inhibition as induced by extreme hypoxia: the possible role of cell cycle arrest as a protection against hypoxia‐induced lethal damage , 1991, Cell proliferation.

[38]  A. Koong,et al.  Therapeutic advantage of hypoxic cells in tumors: a theoretical study. , 1991, Journal of the National Cancer Institute.

[39]  M. Lemmon,et al.  Potentiation by the hypoxic cytotoxin SR 4233 of cell killing produced by fractionated irradiation of mouse tumors. , 1990, Cancer research.

[40]  T S Herman,et al.  Classification of antineoplastic treatments by their differential toxicity toward putative oxygenated and hypoxic tumor subpopulations in vivo in the FSaIIC murine fibrosarcoma. , 1990, Cancer research.

[41]  J. Subjeck,et al.  Resistance to etoposide induced by three glucose-regulated stresses in Chinese hamster ovary cells. , 1989, Cancer research.

[42]  J F Gross,et al.  Morphologic and hemodynamic comparison of tumor and healing normal tissue microvasculature. , 1989, International journal of radiation oncology, biology, physics.

[43]  S. Rockwell,et al.  Randomized clinical trial of mitomycin C as an adjunct to radiotherapy in head and neck cancer. , 1989, International journal of radiation oncology, biology, physics.

[44]  P. Olive,et al.  Use of a carbocyanine dye as a marker of functional vasculature in murine tumours. , 1989, British Journal of Cancer.

[45]  R. Durand Distribution and activity of antineoplastic drugs in a tumor model. , 1989, Journal of the National Cancer Institute.

[46]  C. Grau,et al.  Effect of cancer chemotherapy on the hypoxic fraction of a solid tumor measured using a local tumor control assay. , 1988, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[47]  A. Sartorelli Therapeutic attack of hypoxic cells of solid tumors: presidential address. , 1988, Cancer research.

[48]  Shah-Yukich Aa,et al.  Characterization of solid tumor microvasculature: a three-dimensional analysis using the polymer casting technique. , 1988 .

[49]  C. Bartels,et al.  Coinduction of glucose-regulated proteins and doxorubicin resistance in Chinese hamster cells. , 1987, Proceedings of the National Academy of Sciences of the United States of America.

[50]  D. Chaplin,et al.  Intermittent blood flow in a murine tumor: radiobiological effects. , 1987, Cancer research.

[51]  R. Sutherland,et al.  Induction characteristics of oxygen regulated proteins. , 1986, International journal of radiation oncology, biology, physics.

[52]  D. Chaplin,et al.  Acute hypoxia in tumors: implications for modifiers of radiation effects. , 1986, International journal of radiation oncology, biology, physics.

[53]  J. Henk Late results of a trial of hyperbaric oxygen and radiotherapy in head and neck cancer: a rationale for hypoxic cell sensitizers? , 1986, International journal of radiation oncology, biology, physics.

[54]  P. Fracasso,et al.  Cytotoxicity and DNA lesions produced by mitomycin C and porfiromycin in hypoxic and aerobic EMT6 and Chinese hamster ovary cells. , 1986, Cancer research.

[55]  R. Schimke,et al.  Transient hypoxia enhances the frequency of dihydrofolate reductase gene amplification in Chinese hamster ovary cells. , 1986, Proceedings of the National Academy of Sciences of the United States of America.

[56]  M. Lemmon,et al.  SR-4233: a new bioreductive agent with high selective toxicity for hypoxic mammalian cells. , 1986, International journal of radiation oncology, biology, physics.

[57]  C. Coleman,et al.  Phase I trial of the hypoxic cell radiosensitizer SR-2508: the results of the five to six week drug schedule. , 1986, International journal of radiation oncology, biology, physics.

[58]  T. Grunt,et al.  The vascular pattern of basal cell tumors: light microscopy and scanning electron microscopic study on vascular corrosion casts. , 1985, Microvascular research.

[59]  S. Dische Chemical sensitizers for hypoxic cells: a decade of experience in clinical radiotherapy. , 1985, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[60]  J. Chapman,et al.  The detection and measurement of hypoxic cells in solid tumors , 1984, Cancer.

[61]  J. Subjeck,et al.  Induction of glucose-regulated proteins during anaerobic exposure and of heat-shock proteins after reoxygenation. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[62]  J. Brown,et al.  Clinical trials of radiosensitizers: what should we expect? , 1984, International journal of radiation oncology, biology, physics.

[63]  I. Tannock,et al.  Activity of mitomycin C for aerobic and hypoxic cells in vitro and in vivo. , 1983, Cancer research.

[64]  S. Dische,et al.  Carcinoma of the cervix--anaemia, radiotherapy and hyperbaric oxygen. , 1983, The British journal of radiology.

[65]  R. Mikkelsen Extracellular PH and weak acid/weak base drug uptake and cytotoxicity , 1982 .

[66]  S. Rockwell,et al.  Mitomycin-C as a prototype bioreductive alkylating agent: in vitro studies of metabolism and cytotoxicity. , 1982, International journal of radiation oncology, biology, physics.

[67]  D. M. Brown,et al.  SR-2508: a 2-nitroimidazole amide which should be superior to misonidazole as a radiosensitizer for clinical use. , 1981, International journal of radiation oncology, biology, physics.

[68]  I. Tannock,et al.  Response of Chinese hamster ovary cells to anticancer drugs under aerobic and hypoxic conditions. , 1981, British Journal of Cancer.

[69]  J. Brown,et al.  Partition coefficient as a guide to the development of radiosensitizers which are less toxic than misonidazole. , 1980, Radiation research.

[70]  J M Brown,et al.  Evidence for acutely hypoxic cells in mouse tumours, and a possible mechanism of reoxygenation. , 1979, The British journal of radiology.

[71]  R. Sutherland,et al.  Resistance to adriamycin in multicellular spheroids. , 1979, International journal of radiation oncology, biology, physics.

[72]  R. Hill,et al.  Cell cycle distribution of chronically hypoxic cells and determination of the clonogenic potential of cells accumulated in G2 + M phases after irradiation of a solid tumor in vivo. , 1979, Cancer research.

[73]  J. Hendry,et al.  Radiobiology for the Radiologist , 1979, British Journal of Cancer.

[74]  S. Dische,et al.  Hyperbaric oxygen and radiotherapy: a Medical Research Council trial in carcinoma of the bladder. , 1978, The British journal of radiology.

[75]  A. Rauth,et al.  Differences in the toxicity and metabolism of the 2-nitroimidazole misonidazole (Ro-07-0582) in HeLa and Chinese hamster ovary cells. , 1978, Cancer research.

[76]  J. Brown,et al.  Cytotoxic effects of the hypoxic cell radiosensitizer Ro 7-0582 to tumor cells in vivo. , 1977, Radiation research.

[77]  G. Adams,et al.  Clinical testing of the radiosensitizer Ro 07-0582: experience with multiple doses. , 1977, British Journal of Cancer.

[78]  T. Skovsgaard Transport and binding of daunorubicin, adriamycin, and rubidazone in Ehrlich ascites tumour cells. , 1977, Biochemical pharmacology.

[79]  A. Sartorelli,et al.  Potential Bioreductive Alkylating Agents , 1976 .

[80]  A. Rauth,et al.  Increased cell killing by metronidazole and nitrofurazone of hypoxic compared to aerobic mammalian cells. , 1976, Cancer research.

[81]  J. Brown,et al.  Selective radiosensitization of the hypoxic cells of mouse tumors with the nitroimidazoles metronidazole and Ro 7-0582. , 1975, Radiation research.

[82]  H. Withers,et al.  Enhancement of the radioresponse of a murine tumour by a nitroimidazole , 1975 .

[83]  J. Fowler,et al.  Radiosensitization of C3H Mouse Mammary Tumours by a 2-Nitroimidazole Drug , 1974, British Journal of Cancer.

[84]  P. Sheldon,et al.  Demonstration of radiosensitization of hypoxic cells in solid tumours by metronidazole. , 1974, The British journal of radiology.

[85]  A. Sartorelli,et al.  Potential bioreductive alkylating agents. 1. Benzoquinone derivatives. , 1972, Journal of medicinal chemistry.

[86]  G. Adams,et al.  Radiosensitization of hypoxic cells in vitro: a water-soluble derivative of paranitroacetophenone. , 1972, Nature: New biology.

[87]  J. S. Sawyer Man-made Carbon Dioxide and the “Greenhouse” Effect , 1972, Nature.

[88]  L. H. Gray,et al.  The Histological Structure of Some Human Lung Cancers and the Possible Implications for Radiotherapy , 1955, British Journal of Cancer.

[89]  A. F. Phillips,et al.  Measurement of Electrode Potentials in Living and Dead Tissues , 1954, Nature.

[90]  L. H. Gray,et al.  The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. , 1953, The British journal of radiology.

[91]  H. G. Crabtree,et al.  The Action of Radium on Cancer Cells. II.--Some Factors Determining the Susceptibility of Cancer Cells to Radium , 1933 .

[92]  J. Overgaard,et al.  Pretreatment oxygenation predicts radiation response in advanced squamous cell carcinoma of the head and neck. , 1996, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[93]  S M Bentzen,et al.  Measurement of human tumour oxygenation status by a polarographic needle electrode. An analysis of inter- and intratumour heterogeneity. , 1994, Acta oncologica.

[94]  J. Overgaard Clinical evaluation of nitroimidazoles as modifiers of hypoxia in solid tumors. , 1994, Oncology research.

[95]  R. Durand The influence of microenvironmental factors during cancer therapy. , 1994, In vivo.

[96]  S. Rockwell,et al.  Mitomycin C as an adjunct to postoperative radiation therapy in squamous cell carcinoma of the head and neck: results from two randomized clinical trials. , 1993, International journal of radiation oncology, biology, physics.

[97]  P Vaupel,et al.  Intratumoral pO2 predicts survival in advanced cancer of the uterine cervix. , 1993, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[98]  W. Wilson,et al.  Tumour hypoxia: challenges for cancer chemotherapy , 1992 .

[99]  A. Nias Radiobiology for the Radiologist (3rd Edition) , 1988 .

[100]  A. Nelson,et al.  Characterization of solid tumor microvasculature: a three-dimensional analysis using the polymer casting technique. , 1988, Laboratory investigation; a journal of technical methods and pathology.

[101]  B. Teicher,et al.  The hypoxic tumor cell: a target for selective cancer chemotherapy. , 1980, Biochemical pharmacology.

[102]  T. Matsuzawa,et al.  Tumor regrowth after irradiation; an experimental approach. , 1979, International journal of radiation biology and related studies in physics, chemistry, and medicine.

[103]  G Wiernik,et al.  Hyperbaric oxygen and radiotherapy: a Medical Research Council trial in carcinoma of the cervix. , 1978, The British journal of radiology.

[104]  R. Willson,et al.  Radiosensitization of anoxic cells by metronidazole. , 1973, The British journal of radiology.

[105]  M. Ozima,et al.  Excess Argon in Submarine Basalts and an Earth-Atmosphere Evolution Model , 1972 .

[106]  G. Adams,et al.  Electron-affinic sensitization. I. A structural basis for chemical radiosensitizers in bacteria. , 1969, International Journal of Radiation Biology and Related Studies in Physics Chemistry and Medicine.