Abstract
Tumor angiogenesis is recognized as a major therapeutic target in the fight against cancer. The key involvement of angiogenesis in tumor growth and metastasis has started to redefine chemotherapy and new protocols have emerged. Metronomic chemotherapy, which is intended to prevent tumor angiogenesis, is based on more frequent and low-dose drug administrations compared with conventional chemotherapy. The potential of metronomic chemotherapy was revealed in animal models a decade ago and the efficacy of this approach has been confirmed in the clinic. In the past 5 years, multiple clinical trials have investigated the safety and efficacy of metronomic chemotherapy in a variety of human cancers. While the results have been variable, clinical studies have shown that these new treatment protocols represent an interesting alternative for either primary systemic therapy or maintenance therapy. We review the latest clinical trials of metronomic chemotherapy in adult and pediatric cancer patients. Accumulating evidence suggests that the efficacy of such treatment may not only rely on anti-angiogenic activity. Potential new mechanisms of action, such as restoration of anticancer immune response and induction of tumor dormancy are discussed. Finally, we highlight the research efforts that need to be made to facilitate the optimal development of metronomic chemotherapy.
Key Points
-
Metronomic chemotherapy is based on the chronic administration of chemotherapeutic agents at relatively low, minimally toxic doses, and with no prolonged drug-free breaks
-
Metronomic chemotherapy was originally developed to overcome drug resistance by shifting the therapeutic target from tumor cells to the tumor vasculature
-
In the past decade, several pilot and phase II clinical studies have established the potential efficacy and low toxicity of metronomic chemotherapy in adult and childhood cancer patients
-
Metronomic chemotherapy combined with conventional chemotherapy, radiotherapy and/or targeted therapy is an emerging anti-cancer strategy
-
Recent findings suggest that metronomic chemotherapy may be a multi-targeted cancer therapy rather than a simple anti-angiogenic therapy
-
In addition to inhibiting tumor angiogenesis, metronomic chemotherapy might also restore anticancer immune response and induce tumor dormancy
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Jemal, A. et al. Cancer statistics, 2009. CA Cancer J. Clin. 59, 225–249 (2009).
Folkman, J. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 285, 1182–1186 (1971).
Kerbel, R. S. Tumor angiogenesis. N. Engl. J. Med. 358, 2039–2049 (2008).
Bergers, G. & Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 8, 592–603 (2008).
Jain, R. K. et al. Biomarkers of response and resistance to antiangiogenic therapy. Nat. Rev. Clin. Oncol. 6, 327–338 (2009).
Ebos, J. M. et al. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239 (2009).
Paez-Ribes, M. et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15, 220–231 (2009).
Loges, S., Mazzone, M., Hohensinner, P. & Carmeliet, P. Silencing or fueling metastasis with VEGF inhibitors: antiangiogenesis revisited. Cancer Cell 15, 167–170 (2009).
Kerbel, R. S. & Kamen, B. A. The anti-angiogenic basis of metronomic chemotherapy. Nat. Rev. Cancer 4, 423–436 (2004).
Gasparini, G. Metronomic scheduling: the future of chemotherapy? Lancet Oncol. 2, 733–740 (2001).
Kerbel, R. S. Inhibition of tumor angiogenesis as a strategy to circumvent acquired resistance to anti-cancer therapeutic agents. Bioessays 13, 31–36 (1991).
Klement, G. et al. Continuous low-dose therapy with vinblastine and VEGF receptor-2 antibody induces sustained tumor regression without overt toxicity. J. Clin. Invest. 105, R15–R24 (2000).
Browder, T. et al. Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res. 60, 1878–1886 (2000).
Hanahan, D., Bergers, G. & Bergsland, E. Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J. Clin. Invest. 105, 1045–1047 (2000).
Sarmiento, R. & Gasparini, G. Antiangiogenic metronomic chemotherapy. Onkologie 31, 161–162 (2008).
Colleoni, M. et al. Low-dose oral methotrexate and cyclophosphamide in metastatic breast cancer: antitumor activity and correlation with vascular endothelial growth factor levels. Ann. Oncol. 13, 73–80 (2002).
Colleoni, M. et al. Metronomic low-dose oral cyclophosphamide and methotrexate plus or minus thalidomide in metastatic breast cancer: antitumor activity and biological effects. Ann. Oncol. 17, 232–238 (2006).
Wong, N. S. et al. Phase I/II trial of metronomic chemotherapy with daily dalteparin and cyclophosphamide, twice-weekly methotrexate, and daily prednisone as therapy for metastatic breast cancer using vascular endothelial growth factor and soluble vascular endothelial growth factor receptor levels as markers of response. J. Clin. Oncol. 28, 723–730 (2010).
Gonzalez-Billalabeitia, E. et al. Long-term follow-up of an anthracycline-containing metronomic chemotherapy schedule in advanced breast cancer. Breast J. 15, 551–553 (2009).
Bottini, A. et al. Randomized phase II trial of letrozole and letrozole plus low-dose metronomic oral cyclophosphamide as primary systemic treatment in elderly breast cancer patients. J. Clin. Oncol. 24, 3623–3628 (2006).
Orlando, L. et al. Trastuzumab in combination with metronomic cyclophosphamide and methotrexate in patients with HER-2 positive metastatic breast cancer. BMC Cancer 6, 225 (2006).
Dellapasqua, S. et al. Metronomic cyclophosphamide and capecitabine combined with bevacizumab in advanced breast cancer. J. Clin. Oncol. 26, 4899–4905 (2008).
Garcia-Saenz, J. A. et al. Bevacizumab in combination with metronomic chemotherapy in patients with anthracycline- and taxane-refractory breast cancer. J. Chemother. 20, 632–639 (2008).
Jurado Garcia, J. M. et al. Combined oral cyclophosphamide and bevacizumab in heavily pre-treated ovarian cancer. Clin. Transl. Oncol. 10, 583–586 (2008).
Garcia, A. A. et al. Phase II clinical trial of bevacizumab and low-dose metronomic oral cyclophosphamide in recurrent ovarian cancer: a trial of the California, Chicago, and Princess Margaret Hospital phase II consortia. J. Clin. Oncol. 26, 76–82 (2008).
Glode, L. M., Barqawi, A., Crighton, F., Crawford, E. D. & Kerbel, R. Metronomic therapy with cyclophosphamide and dexamethasone for prostate carcinoma. Cancer 98, 1643–1648 (2003).
Nicolini, A. et al. Oral low-dose cyclophosphamide in metastatic hormone refractory prostate cancer (MHRPC). Biomed. Pharmacother. 58, 447–450 (2004).
Lord, R. et al. Low dose metronomic oral cyclophosphamide for hormone resistant prostate cancer: a phase II study. J. Urol. 177, 2136–2140 (2007).
Fontana, A. et al. Clinical and pharmacodynamic evaluation of metronomic cyclophosphamide, celecoxib, and dexamethasone in advanced hormone-refractory prostate cancer. Clin. Cancer Res. 15, 4954–4962 (2009).
de Weerdt, O. et al. Continuous low-dose cyclophosphamide-prednisone is effective and well tolerated in patients with advanced multiple myeloma. Neth. J. Med. 59, 50–56 (2001).
Buckstein, R. et al. High-dose celecoxib and metronomic “low-dose” cyclophosphamide is an effective and safe therapy in patients with relapsed and refractory aggressive histology non-Hodgkin's lymphoma. Clin. Cancer Res. 12, 5190–5198 (2006).
Kong, D. S. et al. A pilot study of metronomic temozolomide treatment in patients with recurrent temozolomide-refractory glioblastoma. Oncol. Rep. 16, 1117–1121 (2006).
Perry, J. R., Rizek, P., Cashman, R., Morrison, M. & Morrison, T. Temozolomide rechallenge in recurrent malignant glioma by using a continuous temozolomide schedule: the “rescue” approach. Cancer 113, 2152–2157 (2008).
Reardon, D. A. et al. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: a phase II study. Br. J. Cancer 101, 1986–1994 (2009).
Brizzi, M. P. et al. Continuous 5-fluorouracil infusion plus long acting octreotide in advanced well-differentiated neuroendocrine carcinomas. A phase II trial of the Piemonte oncology network. BMC Cancer 9, 388 (2009).
Young, S. D. et al. Phase II clinical trial results involving treatment with low-dose daily oral cyclophosphamide, weekly vinblastine, and rofecoxib in patients with advanced solid tumors. Clin. Cancer Res. 12, 3092–3098 (2006).
Steinbild, S. et al. Metronomic antiangiogenic therapy with capecitabine and celecoxib in advanced tumor patients—results of a phase II study. Onkologie 30, 629–635 (2007).
Kesari, S. et al. Phase II study of metronomic chemotherapy for recurrent malignant gliomas in adults. Neuro. Oncol. 9, 354–363 (2007).
Krzyzanowska, M. K. et al. A phase II trial of continuous low-dose oral cyclophosphamide and celecoxib in patients with renal cell carcinoma. Cancer Chemother. Pharmacol. 60, 135–141 (2007).
Sterba, J. et al. Combined biodifferentiating and antiangiogenic oral metronomic therapy is feasible and effective in relapsed solid tumors in children: single-center pilot study. Onkologie 29, 308–313 (2006).
Kieran, M. W. et al. A feasibility trial of antiangiogenic (metronomic) chemotherapy in pediatric patients with recurrent or progressive cancer. J. Pediatr. Hematol. Oncol. 27, 573–581 (2005).
du Manoir, J. M. et al. Strategies for delaying or treating in vivo acquired resistance to trastuzumab in human breast cancer xenografts. Clin. Cancer Res. 12, 904–916 (2006).
Watanabe, T. et al. Oral uracil and tegafur compared with classic cyclophosphamide, methotrexate, fluorouracil as postoperative chemotherapy in patients with node-negative, high-risk breast cancer: National Surgical Adjuvant Study for Breast Cancer 01 Trial. J. Clin. Oncol. 27, 1368–1374 (2009).
Briasoulis, E. et al. Dose-ranging study of metronomic oral vinorelbine in patients with advanced refractory cancer. Clin. Cancer Res. 15, 6454–6461 (2009).
Nannini, M. et al. To widen the setting of cancer patients who could benefit from metronomic capecitabine. Cancer Chemother. Pharmacol. 64, 189–193 (2009).
Spieth, K., Kaufmann, R. & Gille, J. Metronomic oral low-dose treosulfan chemotherapy combined with cyclooxygenase-2 inhibitor in pretreated advanced melanoma: a pilot study. Cancer Chemother. Pharmacol. 52, 377–382 (2003).
Borne, E. et al. Oral metronomic cyclophosphamide in elderly with metastatic melanoma. Invest. New Drugs doi: 10.1007/s10637-009-9298-5.
Vogt, T. et al. Antiangiogenetic therapy with pioglitazone, rofecoxib, and metronomic trofosfamide in patients with advanced malignant vascular tumors. Cancer 98, 2251–2256 (2003).
Reichle, A. & Vogt, T. Systems biology: a therapeutic target for tumor therapy. Cancer Microenviron. 1, 159–170 (2008).
Kato, H. et al. A randomized trial of adjuvant chemotherapy with uracil-tegafur for adenocarcinoma of the lung. N. Engl. J. Med. 350, 1713–1721 (2004).
Herrlinger, U. et al. UKT-04 trial of continuous metronomic low-dose chemotherapy with methotrexate and cyclophosphamide for recurrent glioblastoma. J. Neurooncol. 71, 295–299 (2005).
Bhatt, R. S. et al. A phase 2 pilot trial of low-dose, continuous infusion, or “metronomic” paclitaxel and oral celecoxib in patients with metastatic melanoma. Cancer 116, 1751–1756 (2010).
Stupp, R. et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).
Yagoda, A., Abi-Rached, B. & Petrylak, D. Chemotherapy for advanced renal-cell carcinoma: 1983–1993. Semin. Oncol. 22, 42–60 (1995).
Bellmunt, J. et al. Activity of a multitargeted chemo-switch regimen (sorafenib, gemcitabine, and metronomic capecitabine) in metastatic renal-cell carcinoma: a phase 2 study (SOGUG-02-06). Lancet Oncol. 11, 350–357 (2010).
Sterba, J., Pavelka, Z. & Slampa, P. Concomitant radiotherapy and metronomic temozolomide in pediatric high-risk brain tumors. Neoplasma 49, 117–120 (2002).
Choi, L. M. et al. Feasibility of metronomic maintenance chemotherapy following high-dose chemotherapy for malignant central nervous system tumors. Pediatr. Blood Cancer 50, 970–975 (2008).
Stempak, D. et al. A pilot pharmacokinetic and antiangiogenic biomarker study of celecoxib and low-dose metronomic vinblastine or cyclophosphamide in pediatric recurrent solid tumors. J. Pediatr. Hematol. Oncol. 28, 720–728 (2006).
Andre, N. et al. Metronomic etoposide/cyclophosphamide/celecoxib regimen given to children and adolescents with refractory cancer: a preliminary monocentric study. Clin. Ther. 30, 1336–1340 (2008).
Le Deley, M. C. et al. High cumulative rate of secondary leukemia after continuous etoposide treatment for solid tumors in children and young adults. Pediatr. Blood Cancer 45, 25–31 (2005).
De Vita, S. et al. Secondary Ph+ acute lymphoblastic leukemia after temozolomide. Ann. Hematol. 84, 760–762 (2005).
Rome, A. et al. Metronomic chemotherapy-induced bilateral subdural hematoma in a child with meningeal carcinomatosis. Pediatr. Blood Cancer 53, 246–247 (2009).
Bocci, G., Nicolaou, K. C. & Kerbel, R. S. Protracted low-dose effects on human endothelial cell proliferation and survival in vitro reveal a selective antiangiogenic window for various chemotherapeutic drugs. Cancer Res. 62, 6938–6943 (2002).
Miller, K. D., Sweeney, C. J. & Sledge, G. W. Jr. Redefining the target: chemotherapeutics as antiangiogenics. J. Clin. Oncol. 19, 1195–1206 (2001).
Pasquier, E., Andre, N. & Braguer, D. Targeting microtubules to inhibit angiogenesis and disrupt tumour vasculature: implications for cancer treatment. Curr. Cancer Drug Targets 7, 566–581 (2007).
Laquente, B., Vinals, F. & Germa, J. R. Metronomic chemotherapy: an antiangiogenic scheduling. Clin. Transl. Oncol. 9, 93–98 (2007).
Dunn, G. P., Bruce, A. T., Ikeda, H., Old, L. J. & Schreiber, R. D. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3, 991–998 (2002).
Zitvogel, L., Apetoh, L., Ghiringhelli, F. & Kroemer, G. Immunological aspects of cancer chemotherapy. Nat. Rev. Immunol. 8, 59–73 (2008).
Kosmaczewska, A., Ciszak, L., Potoczek, S. & Frydecka, I. The significance of Treg cells in defective tumor immunity. Arch. Immunol. Ther. Exp. (Warsz) 56, 181–191 (2008).
Kono, K. et al. CD4(+)CD25high regulatory T cells increase with tumor stage in patients with gastric and esophageal cancers. Cancer Immunol. Immunother. 55, 1064–1071 (2006).
Ghiringhelli, F. et al. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur. J. Immunol. 34, 336–344 (2004).
Loeffler, M., Kruger, J. A. & Reisfeld, R. A. Immunostimulatory effects of low-dose cyclophosphamide are controlled by inducible nitric oxide synthase. Cancer Res. 65, 5027–5030 (2005).
Lutsiak, M. E. et al. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood 105, 2862–2868 (2005).
Banissi, C., Ghiringhelli, F., Chen, L. & Carpentier, A. F. Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model. Cancer Immunol. Immunother. 58, 1627–1634 (2009).
Ghiringhelli, F. et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol. Immunother. 56, 641–648 (2007).
Generali, D. et al. Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients. Clin. Cancer Res. 15, 1046–1051 (2009).
Tanaka, H., Matsushima, H., Mizumoto, N. & Takashima, A. Classification of chemotherapeutic agents based on their differential in vitro effects on dendritic cells. Cancer Res. 69, 6978–6986 (2009).
Tanaka, H., Matsushima, H., Nishibu, A., Clausen, B. E. & Takashima, A. Dual therapeutic efficacy of vinblastine as a unique chemotherapeutic agent capable of inducing dendritic cell maturation. Cancer Res. 69, 6987–6994 (2009).
Man, S. et al. Antitumor effects in mice of low-dose (metronomic) cyclophosphamide administered continuously through the drinking water. Cancer Res. 62, 2731–2735 (2002).
Klement, G. et al. Differences in therapeutic indexes of combination metronomic chemotherapy and an anti-VEGFR-2 antibody in multidrug-resistant human breast cancer xenografts. Clin. Cancer Res. 8, 221–232 (2002).
Huang, J. et al. Vascular remodeling marks tumors that recur during chronic suppression of angiogenesis. Mol. Cancer Res. 2, 36–42 (2004).
Aguirre-Ghiso, J. A. Models, mechanisms and clinical evidence for cancer dormancy. Nat. Rev. Cancer 7, 834–846 (2007).
Udagawa, T. Tumor dormancy of primary and secondary cancers. APMIS 116, 615–628 (2008).
Bergers, G. & Benjamin, L. E. Tumorigenesis and the angiogenic switch. Nat. Rev. Cancer 3, 401–410 (2003).
Meng, S. et al. Circulating tumor cells in patients with breast cancer dormancy. Clin. Cancer Res. 10, 8152–8162 (2004).
Gimbrone, M. A. Jr, Leapman, S. B., Cotran, R. S. & Folkman, J. Tumor dormancy in vivo by prevention of neovascularization. J. Exp. Med. 136, 261–276 (1972).
Brem, S., Brem, H., Folkman, J., Finkelstein, D. & Patz, A. Prolonged tumor dormancy by prevention of neovascularization in the vitreous. Cancer Res. 36, 2807–2812 (1976).
Finkelstein, D. et al. Experimental retinal neovascularization induced by intravitreal tumors. Am. J. Ophthalmol. 83, 660–664 (1977).
Matsuzawa, A., Takeda, Y., Narita, M. & Ozawa, H. Survival of leukemic cells in a dormant state following cyclophosphamide-induced cure of strongly immunogenic mouse leukemia (DL811). Int. J. Cancer 49, 303–309 (1991).
Schirrmacher, V. T-cell immunity in the induction and maintenance of a tumour dormant state. Semin. Cancer Biol. 11, 285–295 (2001).
Mahnke, Y. D., Schwendemann, J., Beckhove, P. & Schirrmacher, V. Maintenance of long-term tumour-specific T-cell memory by residual dormant tumour cells. Immunology 115, 325–336 (2005).
Andre, N. & Pasquier, E. Response to 'Intermittent androgen blockade should be regarded as standard therapy in prostate cancer'. Nat. Clin. Pract. Oncol. 6, E1 (2009).
Seruga, B. & Tannock, I. F. Intermittent androgen blockade should be regarded as standard therapy in prostate cancer. Nat. Clin. Pract. Oncol. 5, 574–576 (2008).
Chuu, C. P., Hiipakka, R. A., Fukuchi, J., Kokontis, J. M. & Liao, S. Androgen causes growth suppression and reversion of androgen-independent prostate cancer xenografts to an androgen-stimulated phenotype in athymic mice. Cancer Res. 65, 2082–2084 (2005).
Sabnis, G. J., Macedo, L. F., Goloubeva, O., Schayowitz, A. & Brodie, A. M. Stopping treatment can reverse acquired resistance to letrozole. Cancer Res. 68, 4518–4524 (2008).
Cabral, F. R. Isolation of Chinese hamster ovary cell mutants requiring the continuous presence of taxol for cell division. J. Cell Biol. 97, 22–29 (1983).
Schibler, M. J. & Cabral, F. Taxol-dependent mutants of Chinese hamster ovary cells with alterations in alpha- and beta-tubulin. J. Cell Biol. 102, 1522–1531 (1986).
Kavallaris, M. et al. Taxol-resistant epithelial ovarian tumors are associated with altered expression of specific beta-tubulin isotypes. J. Clin. Invest. 100, 1282–1293 (1997).
Yang, C. P. et al. A highly epothilone B-resistant A549 cell line with mutations in tubulin that confer drug dependence. Mol. Cancer Ther. 4, 987–995 (2005).
Gatenby, R. A., Silva, A. S., Gillies, R. J. & Frieden, B. R. Adaptive therapy. Cancer Res. 69, 4894–4903 (2009).
Mancuso, P. et al. Circulating endothelial-cell kinetics and viability predict survival in breast cancer patients receiving metronomic chemotherapy. Blood 108, 452–459 (2006).
Bertolini, F., Mancuso, P., Shaked, Y. & Kerbel, R. S. Molecular and cellular biomarkers for angiogenesis in clinical oncology. Drug Discov. Today 12, 806–812 (2007).
Bertolini, F., Mancuso, P., Braidotti, P., Shaked, Y. & Kerbel, R. S. The multiple personality disorder phenotype(s) of circulating endothelial cells in cancer. Biochim. Biophys. Acta 1796, 27–32 (2009).
Hida, A. et al. Nitric oxide acts on the mitochondria and protects human endothelial cells from apoptosis. J. Lab. Clin. Med. 144, 148–155 (2004).
Rigolin, G. M. et al. Neoplastic circulating endothelial cells in multiple myeloma with 13q14 deletion. Blood 107, 2531–2535 (2006).
Hida, K., Hida, Y. & Shindoh, M. Understanding tumor endothelial cell abnormalities to develop ideal anti-angiogenic therapies. Cancer Sci. 99, 459–466 (2008).
Streubel, B. et al. Lymphoma-specific genetic aberrations in microvascular endothelial cells in B-cell lymphomas. N. Engl. J. Med. 351, 250–259 (2004).
Xiong, Y. Q. et al. Human hepatocellular carcinoma tumor-derived endothelial cells manifest increased angiogenesis capability and drug resistance compared with normal endothelial cells. Clin. Cancer Res. 15, 4838–4846 (2009).
Virrey, J. J. et al. Glioma-associated endothelial cells are chemoresistant to temozolomide. J. Neurooncol. 95, 13–22 (2009).
Ohtani, N., Mann, D. J. & Hara, E. Cellular senescence: Its role in tumor suppression and aging. Cancer Sci. 100, 792–797 (2009).
Roninson, I. B. Tumor cell senescence in cancer treatment. Cancer Res. 63, 2705–2715 (2003).
Zetterberg, A. & Larsson, O. Kinetic analysis of regulatory events in G1 leading to proliferation or quiescence of Swiss 3T3 cells. Proc. Natl Acad. Sci. USA 82, 5365–5369 (1985).
Coller, H. A., Sang, L. & Roberts, J. M. A new description of cellular quiescence. PLoS Biol. 4, e83 (2006).
Goymer, P. Natural selection: The evolution of cancer. Nature 454, 1046–1048 (2008).
Meiler, J. & Schuler, M. Therapeutic targeting of apoptotic pathways in cancer. Curr. Drug Targets 7, 1361–1369 (2006).
Perez-Tomas, R. Multidrug resistance: retrospect and prospects in anti-cancer drug treatment. Curr. Med. Chem. 13, 1859–1876 (2006).
Merlo, L. M., Pepper, J. W., Reid, B. J. & Maley, C. C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 6, 924–935 (2006).
Andre, N. & Pasquier, E. For cancer, seek and destroy or live and let live? Nature 460, 324 (2009).
Marusyk, A. & Polyak, K. Tumor heterogeneity: Causes and consequences. Biochim. Biophys. Acta 1805, 105–117 (2010).
Acknowledgements
E. Pasquier and M. Kavallaris are supported by the Children's Cancer Institute Australia for Medical Research, which is affiliated with the University of New South Wales and Sydney Children's Hospital. E. Pasquier is supported by an “Early Career Development” Fellowship from the Cancer Institute New South Wales and a grant from the “Young Researchers Fund” from the Balnaves Foundation. M. Kavallaris is supported by an Australian Government, National Health and Medical Research Council (NHMRC) Senior Research Fellowship, NHMRC Project Grants and Cancer Council New South Wales Program Grant.
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing financial interests.
Rights and permissions
About this article
Cite this article
Pasquier, E., Kavallaris, M. & André, N. Metronomic chemotherapy: new rationale for new directions. Nat Rev Clin Oncol 7, 455–465 (2010). https://doi.org/10.1038/nrclinonc.2010.82
Published:
Issue Date:
DOI: https://doi.org/10.1038/nrclinonc.2010.82
This article is cited by
-
Impact of metronomic trabectedin combined with low-dose cyclophosphamide on sarcoma microenvironment and correlation with clinical outcome: results from the TARMIC study
Molecular Cancer (2024)
-
Reshaping the tumor microenvironment of cold soft-tissue sarcomas with oncolytic viral therapy: a phase 2 trial of intratumoral JX-594 combined with avelumab and low-dose cyclophosphamide
Molecular Cancer (2024)
-
Targeting CXCL16 and STAT1 augments immune checkpoint blockade therapy in triple-negative breast cancer
Nature Communications (2023)
-
CHEOPS trial: a GINECO group randomized phase II assessing addition of a non-steroidal aromatase inhibitor to oral vinorelbine in pre-treated metastatic breast cancer patients
Breast Cancer (2023)
-
Randomized phase 2 trial of intravenous oncolytic virus JX-594 combined with low-dose cyclophosphamide in patients with advanced soft-tissue sarcoma
Journal of Hematology & Oncology (2022)