Skip to main content

Overview of Resistance to Systemic Therapy in Patients with Breast Cancer

  • Chapter
Breast Cancer Chemosensitivity

Abstract

Breast cancer is the most common cancer and the second leading cause of cancer death in American women. It was the second most common cancer in the world in 2002, with more than 1 million new cases. Despite advances in early detection and the understanding of the molecular bases of breast cancer biology, about 30% of patients with early-stage breast cancer have recurrent disease. To offer more effective and less toxic treatment, selecting therapies requires considering the patient and the clinical and molecular characteristics of the tumor. Systemic treatment of breast cancer includes cytotoxic, hormonal, and immunotherapeutic agents. These medications are used in the adjuvant, neoadjuvant, and metastatic settings. In general, systemic agents are active at the beginning of therapy in 90% of primary breast cancers and 50% of metastases. However, after a variable period of time, progression occurs. At that point, resistance to therapy is not only common but expected. Herein we review general mechanisms of drug resistance, including multidrug resistance by P-glyucoprotein and the multidrug resistance protein family in association with specific agents and their metabolism, emergence of refractory tumors associated with multiple resistance mechanisms, and resistance factors unique to host-tumor-drug interactions. Important anticancer agents specific to breast cancer are described.

Breast cancer is the most common type of cancer and the second leading cause of cancer death in American women. In 2002, 209,995 new cases of breast cancer were registered, and 42,913 patients died of it.1 In 5 years, the annual prevalence of breast cancer will reach 968,731 cases in the United States.2 World wide, the problem is just as significant, as breast cancer is the most frequent cancer after nonmelanoma skin cancer, with more than 1 million new cases in 2002 and an expected annual prevalence of more than 4.4 million in 5 years.1

Breast cancer treatment currently requires the joint efforts of a multidisciplinary team. The alternatives for treatment are constantly expanding. With the use of new effective chemotherapy, hormone therapy, and biological agents and with information regarding more effective ways to integrate systemic therapy, surgery, and radiation therapy, elaborating an appropriate treatment plan is becoming more complex. Developing such a plan should be based on knowledge of the benefits and potential acute and late toxic effects of each of the therapy regiments.

Despite advances in early detection and understanding of the molecular bases of breast cancer biology, approximately 30% of all patients with early-stage breast cancer have recurrent disease, which is metastatic in most cases.3 The rates of local and systemic recurrence vary within different series, but in general, distant recurrences are dominant, strengthening the hypothesis that breast cancer is a systemic disease from presentation. On the other hand, local recurrence may signal a posterior systemic relapse in a considerable number of patients within 2 to 5 years after completion of treatment.4

To offer better treatment with increased efficacy and low toxicity, selecting therapies based on the patient and the clinical and molecular characteristics of the tumor is necessary. Consideration of these factors should be incorporated in clinical practice after appropriate validation studies are performed to avoid confounding results, making them true prognostic and predictive factors.5 A prognostic factor is a measurable clinical or biological characteristic associated with a disease-free or overall survival period in the absence of adjuvant therapy, whereas a predictive factor is any measurable characteristic associated with a response or lack of a response to a specific treatment.6 The main prognostic factors associated with breast cancer are the number of lymph nodes involved, tumor size, histological grade, and hormone receptor status, the first two of which are the basis for the AJCC staging system. The sixth edition of the American Joint Committee on Cancer staging system allows better prediction of prognosis by stage.7 However, after determining the stage, histological grade, and hormone receptor status, the tumor can behave in an unexpected manner, and the prognosis can vary. Other prognostic and predictive factors have been studied in an effort to explain this phenomenon, some of which are more relevant than others: HER-2/neu gene amplification and protein expression,8,9 expression of other members of the epithelial growth factor receptor family,10,11 S phase fraction, DNA ploidy,12 p53 gene mutations,13 cyclin E,14 p27 dysregulation,15 the presence of tumor cells in the circulation16 or bone marrow,17 and perineural and lymphovascular space invasion.18

Systemic treatment of breast cancer includes the use of cytotoxic, hormonal, and immunotherapeutic agents. All of these agents are used in the adjuvant, neoadjuvant, and metastatic setting. Adjuvant systemic therapy is used in patients after they undergo primary surgical resection of their breast tumor and axillary nodes and who have a significant risk of systemic recurrence. Multiple studies have demonstrated that adjuvant therapy for early-stage breast cancer produces a 23% or greater improvement in disease-free survival and a 15% or greater increase in overall survival rates.19 Recommendations for the use of adjuvant therapy are based on the individual patient’s risk and the balance between absolute benefit and toxicity. Anthracycline-based regimens are preferred, and the addition of taxanes increases the survival rate in patients with lymph node-positive disease.20 Adjuvant hormone therapy accounts for almost two thirds of the benefit of adjuvant therapy overall in patients with hormone-receptor-positive breast cancer.21 Tamoxifen is considered the standard of care in premenopausal patients.22 In comparison, the aromatase inhibitor anastrozole has been proven to be superior to tamoxifen in postmenopausal patients with early-stage breast cancer.23 The adjuvant use of monoclonal antibodies and targeted therapies other than hormone therapy is being studied. Interestingly, some patients have an early recurrence even though they have a tumor with good prognostic features and at a favorable stage. These recurrences have been explained by the existence of certain cellular characteristics at the molecular level that make the tumor cells resistant to therapy. Selection of resistant cell clones of micrometastatic disease has also been proposed as an explanation for these events.24,25

Neoadjuvant systemic therapy, which is the standard of care for patients with locally advanced and inflammatory breast cancer, is becoming more popular. It reduces the tumor volume, thus increasing the possibility of breast conservation, and at the same time allows identification of in vivo tumor sensitivity to different agents.26 The pathological response to neoadjuvant systemic therapy in the breast and lymph nodes correlates with patient survival.27,28 Use of this treatment modality produces survival rates identical to those obtained with the standard adjuvant approach.29 The rates of pathological complete response (pCR) to neoadjuvant systemic therapy vary according to the regimen used, ranging from 6% to 15% with anthracycline-based regimens30,31 to almost 30% with the addition of a noncross-resistant agent such as a taxane.32,33 In one study, the addition of neoadjuvant trastuzumab in patients with HER-2-positive breast tumors increased the pCR rate to 65%.34 Primary hormone therapy has also been used in the neoadjuvant systemic setting. Although the pCR rates with this therapy are low, it significantly increases breast conservation.35,36 Currently, neoadjuvant systemic therapy is an important tool in not only assessing tumor response to an agent but also studying the mechanisms of action of the agent and its effects at the cellular level. However, no tumor response is observed in some cases despite the use of appropriate therapy. The tumor continues growing during treatment in such cases, a phenomenon called primary resistance to therapy.37

The use of palliative systemic therapy for metastatic breast cancer is challenging. Five percent of newly diagnosed cases of breast cancer are metastatic, and 30% of treated patients have a systemic recurrence.2,3,38 Once metastatic disease develops, the possibility of a cure is very limited or practically nonexistent. In this heterogeneous group of patients, the 5-year survival rate is 20%, and the median survival duration varies from 12 to 24 months.39 In this setting, breast cancer has multiple clinical presentations, and the therapy for it should be chosen according to the patient’s tumor characteristics, previous treatment, and performance status with the goal of improving survival without compromising quality of life. Treatment resistance is most commonly seen in such patients. They initially may have a response to different agents, but the responses are not sustained, and, in general, the rates of response to subsequent agents are lower. Table 1 summarizes metastatic breast cancer response rates to single-agent systemic therapy.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Anonymous. Cancer Incidence, Mortality and Prevalence Worldwide, Version 1.0. GLOBOCAN: IARC Press, 2002, (http//:www-dep.iarc.fr/daba/infodata.htm.).

    Google Scholar 

  2. Jemal A, Murray T, Samuels A et al. Cancer statistics, 2003. CA Cancer J Clin 2003; 53:5–26.

    Article  PubMed  Google Scholar 

  3. Pisani P, Bray F, Parkim DN. Estimates of the worldwide prevalence of cancer for 25 sites in the adult population. Int J Cancer 2002; 97:72–81.

    Article  PubMed  CAS  Google Scholar 

  4. Collyar DE. Breast cancer; a global prespective. J Clin Oncol 2002; 19(18 Suppl):101S–105S.

    Google Scholar 

  5. McGuire WL. Breast cancer prognostic factors: Evaluation guidelines. J Natl Cancer Inst 1991; 83:154–155.

    Article  PubMed  CAS  Google Scholar 

  6. Winer EP, Morrow M, Osborne CK et al. Malignat tumors of the breast. In: DeVita VT, Hellman S, Rosenberg S, eds. Cancer Principles and Practices of Oncology, chap 37.2. Philadephia, PA: 2001:1651–1717.

    Google Scholar 

  7. Singletary SE, Allred C, Ashley P et al. Revision of the American Joint Committee on Cancer staging system for breast cancer. J Clin Oncol 2002; 20:3628–3636.

    Article  PubMed  Google Scholar 

  8. Slamon DJ, Clark GM, Wong SG et al. Human breast cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987; 235:177–182.

    Article  PubMed  CAS  Google Scholar 

  9. Tommasi S, Paradiso A, Mangia A et al. Biological correlation between HER-2/neu and proliferative activity in human breast cancer. Anticancer Res 1991; 11:1395–1400.

    PubMed  CAS  Google Scholar 

  10. Fox SB, Leek RD, Smith K et al. Tumor angiogenesis in node-negative breast carcinomas — Relationship with epidermal growth factor receptor, estrogen receptor, and survival. Breast Cancer Res Treat 1994; 29:109–116.

    Article  PubMed  CAS  Google Scholar 

  11. Gasparini G, Boracchi P, Bevilacqua P et al. A multiparametric study on the prognostic value of epidermal growth factor receptor in operable breast carcinoma. Breast Cancer Res Treat 1994; 29:59–71.

    Article  PubMed  CAS  Google Scholar 

  12. Hedley DW, Clark GM, Cornelisse CJ et al. Consensus review of the clinical utility of DNA cytometry in carcinoma of the breast. Report of the DNA Cytometry Consensus Conference. Cytometry 1993; 14:482–485.

    Article  PubMed  CAS  Google Scholar 

  13. Makris A, Powles TJ, Dowsett M et al. p53 protein overexpression and chemosensitivity in breast cancer. The Lancer 1995; 345:1181–1182.

    Article  CAS  Google Scholar 

  14. Keyomarsi K, Tucker SL, Buchholz TA et al. Cyclin E and, survival in patients with breast cancer. New England Journal of Medicine Online 2002; 347:1566–1575.

    Article  CAS  Google Scholar 

  15. Alkarain A, Slingerland J. Deregulation of p27 by oncogenic signaling and its prognostic significance in breast cancer. Breast Cancer Res 2004; 6:13–21.

    Article  PubMed  CAS  Google Scholar 

  16. Cristofanilli M, Budd GT, Ellis MJ et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 2004; 351:781–791.

    Article  PubMed  CAS  Google Scholar 

  17. Funke IM, Zia A, Wild C et al. Phenotype of disseminated tumor cells in bone marrow of breast cancer patients. J Clin Oncol 2001; 7:3670S.

    Google Scholar 

  18. Gasparini G, Weidner N, Bevilacqua P et al. Tumor microvessel density, p53 expression, tumor size, and peritumoral lymphatic vessel invasion are relevant prognostic markers in node-negative breast carcinoma. J Clin Oncol 1994; 12:454–466.

    PubMed  CAS  Google Scholar 

  19. Anonymous. The national institutes of health consensus development conference: Adjuvant therapy for breast cancer. Bethesda, Maryland, USA. November 1–3, 2000. Proceedings. J Natl Cancer Inst Monogr 2001; 1–152.

    Google Scholar 

  20. Henderson IC, Berry DA, Demetri GD et al. Improved outcomes from adding sequential Paclitaxel but not from escalating Doxorubicin dose in an adjuvant chemotherapy regimen for patients with node-positive primary breast cancer. J Clin Oncol 2003; 21:976–983.

    Article  PubMed  CAS  Google Scholar 

  21. Early Breast Cancer Trialists’ Collaborative Group. Tamoxifen for early breast cancer: An overview of the randomised trials. Lancet 1998; 351:1451–1467.

    Article  Google Scholar 

  22. Early Breast Cancer, Trialists’ Collaborative Group. Tamoxifen for early breast cancer. Cochrane Database Syst Rev 2001; 1:CD000486.

    Google Scholar 

  23. Buzdar AU. Data from the Arimidex, tamoxifen, alone or in combination (ATAC) trial: Implications for use of aromatase inhibitors in 2003. Clin Cancer Res 2004; 10:355S–361S.

    Article  PubMed  CAS  Google Scholar 

  24. Haq R, Zanke B. Inhibition of apoptotic signaling pathways in cancer cells as a mechanism of chemotherapy resistance. Cancer Metastasis Rev 1998; 17:233–239.

    Article  PubMed  CAS  Google Scholar 

  25. DeVita Jr VT. The James Ewing lecture. The relationship between tumor mass and resistance to chemotherapy. Implications for surgical adjuvant treatment of cancer. Cancer 1983; 51:1209–1220.

    Article  PubMed  Google Scholar 

  26. Green M, Hortobagyi GN. Neoadjuvant chemotherapy for operable breast cancer. Oncology (Huntingt) 2002; 16:871–84, (889).

    Google Scholar 

  27. Fisher B, Bryant J, Wolmark N et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 1998; 16:2672–2685.

    PubMed  CAS  Google Scholar 

  28. Kuerer HM, Newman LA, Buzdar AU et al. Pathologic tumor response in the breast following neoadjuvant chemotherapy predicts axillary lymph node status. Cancer Journal From Scientific American 1998; 4:230–236.

    PubMed  CAS  Google Scholar 

  29. Fisher B, Bryant J, Wolmark N et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 1998; 16:2672–2685.

    PubMed  CAS  Google Scholar 

  30. Fisher B, Bryant J, Wolmark N et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 1998; 16:2672–2685.

    PubMed  CAS  Google Scholar 

  31. Bear HD, Anderson S, Brown A et al. The effect on tumor response of adding sequential preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: Preliminary results from National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol 2003; 21:4165–4174.

    Article  PubMed  CAS  Google Scholar 

  32. Bear HD, Anderson S, Brown A et al. The effect on tumor response of adding sequential preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: Preliminary results from National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol 2003; 21:4165–4174.

    Article  PubMed  CAS  Google Scholar 

  33. Heys SD, Hutcheon AW, Sarkar TK et al. Neoadjuvant docetaxel in breast cancer: 3-year survival results from the Aberdeen trial. Clin Breast Cancer 2002; 3(Suppl 2):S69–S74.

    Article  PubMed  Google Scholar 

  34. Buzdar AU, Hunt KK, Smith T et al. Significantly higher pathological complete remission (PCR) rate following neoadjuvant therapy with trastuzumab (H), paclitaxel (P), and anthracycline-containing chemotherapy (CT): Initial results of a randomized trial in operable breast cancer (BC) with HER/ 2 positive disease. Proc Am Soc Clin Oncol 2004; 22(14S).

    Google Scholar 

  35. Ellis MJ, Rosen E, Dressman H et al. Neoadjuvant comparisons of aromatase inhibitors and tamoxifen: Pretreatment determinants of response and on-treatment effect. J Steroid Biochem Mol Biol 2003; 86:301–307.

    Article  PubMed  CAS  Google Scholar 

  36. Huober J, Krainick-Strobel U, Kurek R et al. Neoadjuvant endocrine therapy in primary breast cancer. Clin Breast Cancer 2004;5:341–347.

    Article  PubMed  CAS  Google Scholar 

  37. Muggia FM. Primary chemotherapy: Concepts and issues. Prog Clin Biol Res 1985; 201:377–383.

    PubMed  CAS  Google Scholar 

  38. Parkin DM, Bray F, Ferlay J et al. Estimating the world cancer burden. Globocan 2000. Int J Cancer 2001; 94:153–156.

    Article  PubMed  CAS  Google Scholar 

  39. Cardoso F, Di Leo A, Lohrisch C et al. Second and subsequent lines of chemotherapy for metastatic breast cancer: What did we learn in the last two decades?. Annals Oncol 2002; 3:197–207.

    Article  Google Scholar 

  40. Thomas E, Holmes FA, Smith TL et al. The use of alternate, noncross-resistant adjuvant chemotherapy on the basis of pathologic response to a neoadjuvant doxorubicin-based regimen in women with operable breast cancer: Long-term results from a prospective randomized trial. J Clin Oncol 2004; 22:2294–2302.

    Article  PubMed  CAS  Google Scholar 

  41. Strumberg D, Nitiss JL, Rose A et al. Mutation of a conserved serine residue in a quinolone-resistant type II topoisomerase alters the enzyme-DNA and drug interactions. J Biol Chem 1999; 274:7292–7301.

    Article  PubMed  CAS  Google Scholar 

  42. Biedler JL, Riehm H. Cellular resistance to actinomycin D in Chinese hamster cells in vitro: Cross-resistance, radioauto graphic, and cytogenetic studies. Cancer Res 1970; 30:1174–1184.

    PubMed  CAS  Google Scholar 

  43. Riordan JR, Ling V. Genetic and biochemical characterization of multidrug, resistance. Pharmacol Ther 1985; 28:51–75.

    Article  PubMed  CAS  Google Scholar 

  44. Biedler JL, Riehm H. Cellular resistance to actinomycin D in Chinese hamster cells in vitro: Cross-resistance, radioauto graphic, and cytogenetic studies. Cancer Res 1970; 30:1174–1184.

    PubMed  CAS  Google Scholar 

  45. Gros P, Croop J, Housman D. Mammalian multidrug resistance gene: Complete cDNA sequence indicates strong homology to bacterial transport proteins. Cell 1986; 47:371–380.

    Article  PubMed  CAS  Google Scholar 

  46. Volk EL, Rohde K, Rhee M et al. Metho trexate cross-resistance in a mitoxantrone-selected multidrug-resistant MCF7 breast cancer cell line is attributable to enhanced energy-dependent drug efflux. Cancer Res 2000; 60:3514–3521.

    PubMed  CAS  Google Scholar 

  47. Izquierdo MA, Scheffer GL, Flens MJ et al. Relationship of LRP-human major vault protein to in vitro and clinical resistance to anticancer drugs. Cytotechnology 1996; 19:191–197.

    Article  PubMed  CAS  Google Scholar 

  48. Vassetzky YS, Alghisi GC, Gasser SM. DNA topoisomerase II mutations and resistance to anti-tumor drugs. Bioessays 1995; 17:767–774.

    Article  PubMed  CAS  Google Scholar 

  49. Fernandes DJ, Qiu J, Catapano CV. DNA topoisomerase II isozymes involved in anticancer drug action and resistance. Adv Enzyme Regul 1995; 35:265–281.

    Article  PubMed  CAS  Google Scholar 

  50. Chen YN, Mickley LA, Schwartz AM et al. Characterization of adriamycin-resistant human breast cancer cells which display overexpression of a novel resistance-related membrane protein. J Biol Chem 1990; 265:10073–10080.

    PubMed  CAS  Google Scholar 

  51. Safa AR, Glover CJ, Meyers MB et al. Vinblastine, photoaffinity labeling of a high molecular weight surface membrane glycoprotein specific for multidrug-resistant cells. J Biol Chem 1986; 261:6137–6140.

    PubMed  CAS  Google Scholar 

  52. Choi KH, Chen CJ, Kriegler M et al. An altered pattern of cross-resistance in multidrug-resistant human cells results from spontaneous mutations in the mdrl (P-glycoprotein) gene. Cell 1988. 53:519–529.

    Article  PubMed  CAS  Google Scholar 

  53. Lee K, Belinsky MG, Bell DW et al. Isolation of MOAT-B, a widely expressed multidrug resistance-associated protein/canalicular multispecific organic anion transporter-related transporter. Cancer Res 1998; 58:2741–2747.

    PubMed  CAS  Google Scholar 

  54. Zhan Z, Sandor VA, Gamelin E et al. Expression of the multidrug, resistance-associated protein gene in refractory lymphoma: Quantitation by a validated polymerase chain reaction assay. Blood 1997; 89:3795–3800.

    PubMed  CAS  Google Scholar 

  55. Zhang H, D’Arpa P, Liu LF. A model for tumor cell killing by topoisomerase poisons. Cancer Cell 1990; 2:23–27.

    CAS  Google Scholar 

  56. Hochhauser D, Harris AL. The role of topoisomerase II alpha and beta, in drug resistance. Cancer Treat Rev 1993; 19:181–194.

    Article  PubMed  CAS  Google Scholar 

  57. Drake FH, Hofmann GA, Bartus HF et al. Biochemical and pharmacological properties of p170 and p180 forms of topoisomerase II. Biochemistry 1989; 28:8154–8160.

    Article  PubMed  CAS  Google Scholar 

  58. Larsen AK, Skladanowski A. Cellular resistance to topoisomerase-targeted drugs: From drug uptake to cell death. Biochim Biophys Acta 1998; 1400:257–274.

    PubMed  CAS  Google Scholar 

  59. Larsen AK, Skladanowski A. Cellular resistance to topoisomerase-targeted drugs: From drug uptake to cell death. Biochim Biophys Acta 1998; 1400:257–274.

    PubMed  CAS  Google Scholar 

  60. Matsumoto Y, Takano H, Fojo T. Cellular adaptation to drug exposure: Evolution of the drug-resistant phenotype. Cancer Res 1997; 57:5086–5092.

    PubMed  CAS  Google Scholar 

  61. Liu LF. DNA topoisomerase poisons as antitumor drugs. Annu Rev Biochem 1989; 58:351–375.

    Article  PubMed  CAS  Google Scholar 

  62. Giovanella BC, Stehlin JS, Wall ME et al. DNA topoisomerase I—targeted chemotherapy of human colon cancer in xenografts. Science 1989; 246:1046–1048.

    Article  PubMed  CAS  Google Scholar 

  63. Mannervik B, Danielson UH. Glutathione transferases—structure and catalytic activity. CRC Crit Rev Biochem 1988; 23:283–337.

    Article  PubMed  CAS  Google Scholar 

  64. Hayes JD, Pulford DJ. The glutathione S-transferase supergene family: Regulation of GST and the contribution of the isoenzymes to cancer chemoprotection, and drug resistance. Crit Rev Biochem Mol Biol 1995; 30:445–600.

    Article  PubMed  CAS  Google Scholar 

  65. Brix LA, Nicoll R, Zhu X et al. Structural and functional characterisation of human sulfotransferases. Chem Biol Interact 1998; 109:123–127.

    Article  PubMed  CAS  Google Scholar 

  66. Ivy SP, Tulpule A, Fairchild, CR et al. Altered regulation of P-450IA1 expression in a multidrug-resistant MCF-7 human breast cancer cell line. J Biol Chem 1988; 263:19119–19125.

    PubMed  CAS  Google Scholar 

  67. Thorgeirsson SS, Huber BE, Sorrel S et al. Expression of the multidrug-resistant gene in hepatocarcin ogenesis and regenerating rat liver Science 1987; 236:1120–1122.

    Article  PubMed  CAS  Google Scholar 

  68. Schecter RL, Alaoui-Jamali MA, Woo A et al. Expression of a rat glutathione-S-transferase complementary DNA in rat mammary carcinoma cells: Impact upon alkylator-induced toxicity. Cancer Res 1993; 53:4900–4906.

    PubMed  CAS  Google Scholar 

  69. Leyland-Jones BR, Townsend AJ, Tu CP et al. Antineoplastic drug sensitivity of human MCF-7 breast cancer cells stably transfected with a human alpha class glutathione S-transferase gene. Cancer Res 1991; 51:587–594.

    PubMed  CAS  Google Scholar 

  70. Berhane K, Hao XY, Egyhazi S et al. Contribution of glutathione transferase M3-3 to 1,3-bis (2-chloroethyl)-1-nitrosoureas resistance in a human nonsmall cell lung cancer cell line. Cancer Res 1993; 53:4257–4261.

    PubMed  CAS  Google Scholar 

  71. Awasthi S, Singhal SS, Srivastava SK et al. Adenosine triphosphate-dependent transport of doxorubicin, daunomycin, and vinblastine in human tissues by a mechanism distinct from the P-glycoprotein. J Clin Invest 1994; 93:958–965.

    Article  PubMed  CAS  Google Scholar 

  72. Jedlitschky G, Leier I, Buchholz U et al. ATP-dependent transport of glutathione S-conjugates by the multidrug resistance-associated protein. Cancer Res 1994; 54:4833–4836.

    PubMed  CAS  Google Scholar 

  73. Muller M, Meijer C, Zaman GJ et al. Overexpression of the gene encoding the multidrug resistance-associated protein results in increased ATP-dependent glutathione S-conjugate transport. Proc Natl Acad Sci USA 1994; 91:13033–13037.

    Article  PubMed  CAS  Google Scholar 

  74. Sinha BK, Mimnaugh EG, Rajagopalan S et al. Adriamycin activation and oxygen free radical formation in human breast tumor cells: Protective role of glutathione peroxidase in adriamycin resistance. Cancer Res 1989; 49:3844–3848.

    PubMed  CAS  Google Scholar 

  75. Sinha BK. Free radicals in anticancer drug pharmacology. Chem Biol Interact 1989; 69:293–317.

    Article  PubMed  CAS  Google Scholar 

  76. Hall AG. Review: The role of glutathione in the regulation of apoptosis. Eur J Clin Invest 1999; 29:238–245.

    Article  PubMed  CAS  Google Scholar 

  77. Bellamy CO. p53 and apoptosis. Br Med Bull 1997; 53:522–538.

    PubMed  CAS  Google Scholar 

  78. Reed JC, Miyashita T, Takayama S et al. BCL-2 family proteins: Regulators of cell death involved in the pathogenesis of cancer and resistance to therapy. Journal of Cellular Biochemistry 1996; 60:23–32.

    Article  PubMed  CAS  Google Scholar 

  79. Reed JC. Bcl-2 family proteins: Regulators of apoptosis and chemoresistance in hematologic malignancies. Semin Hematol 1997; 34:9–19.

    PubMed  CAS  Google Scholar 

  80. Poplack DG, Reaman G. Acute lymphoblastic leukemia in childhood,. Pediatr Clin North Am 1988; 35:903–932.

    PubMed  CAS  Google Scholar 

  81. Chen G, Jaffrezou JP, Fleming WH et al. Prevalence of multidrug resistance related to activation of the mdrl gene in human sarcoma mutants derived by single-step doxorubicin selection. Cancer Res 1994; 54:4980–4987.

    PubMed  CAS  Google Scholar 

  82. Laredo J, Huynh A, Muller C et al. Effect of the protein kinase C inhibitor staurosporine on chemosensitivity to daunorubicin of normal and leukemic fresh myeloid cells. Blood 1994; 84:229–237.

    PubMed  CAS  Google Scholar 

  83. van der Kolk DM, de Vries EG, Koning JA et al. Activity and expression of the multidrug resistance proteins MRP1 and MRP2 in acute myeloid leukemia cells, tumor cell lines, and normal hematopoietic CD34+ peripheral blood cells. Clin Cancer Res 1998; 4:1727–1736.

    PubMed  Google Scholar 

  84. Chernov MV, Stark GR. The p53 activation and apoptosis induced by DNA damage are reversibly inhibited by salicylate. Oncogene 1997; 14:2503–2510.

    Article  PubMed  Google Scholar 

  85. Tanizawa A, Kubota M, Takimoto T et al. Prevention of adriamycin-induced interphase death by 3-aminobenzamide and nicotinamide in a human promyelocytic leukemia cell line. Biochem Biophys Res Commun 1987; 144:1031–1036.

    Article  PubMed  CAS  Google Scholar 

  86. Larsen AK, Skladanowski A. Cellular resistance to topoisomerase-targeted drugs: From drug uptake to cell death. Biochim Biophys Acta 1998; 1400:257–274.

    PubMed  CAS  Google Scholar 

  87. Withoff S, de Jong S, de Vries EG et al. Human DNA topoisomerase II: Biochemistry and role in chemotherapy resistance (review). Anticancer Res 1996; 16:1867–1880.

    PubMed  CAS  Google Scholar 

  88. Withoff S, de Jong S, de Vries EG et al. Human DNA topoisomerase II: Biochemistry and role in chemotherapy resistance (review). Anticancer Res 1996; 16:1867–1880.

    PubMed  CAS  Google Scholar 

  89. Finlay GJ, Baguley BC, Snow K et al. Multiple patterns of resistance of human leukemia cell sublines to amsacrine analogues. J Natl Cancer Inst 1990; 82:662–667.

    Article  PubMed  CAS  Google Scholar 

  90. Seidman AD, Reichman BS, Crown JP et al. Paclitaxel as second and subsequent therapy for metastatic breast cancer: Activity independent of prior anthracycline response. J Clin Oncol 1995; 13:1152–1159.

    PubMed  CAS  Google Scholar 

  91. Wilson WH, Berg SL, Bryant G et al. Paclitaxel in doxorubicin-refractory or mitoxantrone-refractory breast cancer: A phase I/II trial of 96-hour infusion. J Clin Oncol 1994; 12:1621–1629.

    PubMed  CAS  Google Scholar 

  92. Anderson H, Hopwood P, Prendiville J et al. A randomised study of bolus vs continuous pump infusion of ifosfamide and doxorubicin with oral etoposide for small cell lung cancer. Br J Cancer 1993; 67:1385–1390.

    PubMed  CAS  Google Scholar 

  93. Wilson L, Miller HP, Farrell KW et al. Taxol stabilization microtubules in vitro: Dynamics of tubulin addition and loss at opposite microtubule ends. Biochemistry 1985; 24:5254–5262.

    Article  PubMed  CAS  Google Scholar 

  94. Sparreboom A, van Tellingen O, Nooijen WJ et al. Preclinical pharmacokinetics of paclitaxel and docetaxel. Anticancer Drugs 1998; 9:1–17.

    Article  PubMed  CAS  Google Scholar 

  95. Greenberger LM, Williams SS, Horwitz SB. Biosynthesis of heterogeneous forms of multidrug resistance-associated glycoproteins. J Biol Chem 1987; 262:13685–13689.

    PubMed  CAS  Google Scholar 

  96. Tolcher AW, Cowan KH, Solomon D et al. Phase I crossover study of paclitaxel with r-verapamil in patients with metastatic breast cancer. J Clin Oncol 1996; 14:1173–1184.

    PubMed  CAS  Google Scholar 

  97. Horwitz SB, Lothstein L, Manfredi JJ et al. Taxol: Mechanisms of action and resistance. Annals of the New York Academy of Sciences 1986; 466:733–744.

    Article  PubMed  CAS  Google Scholar 

  98. Torre K, Horwitz SB. Mechanisms of Taxol-induced cell death are concentration dependent. Cancer Research 1998; 58:3620–3626.

    Google Scholar 

  99. Allegra CJ, Chabner BA, Drake JC et al. Enhanced inhibition of thymidylate synthase by methotrexate polyglutamates. J Biol Chem 1985; 260:9720–9726.

    PubMed  CAS  Google Scholar 

  100. Priest DG, Ledford BE, Doig MT. Increased thymidylate synthetase in 5-fluorodeoxyuridine resistant cultured hepatoma cells. Biochem Pharmacol 1980; 29:1549–1553.

    Article  PubMed  CAS  Google Scholar 

  101. Bapat AR, Zarow C, Danenberg PV. Human luekemic cells resitant to 5-fluoro-2′-deoxyuridine contain a thymidylate synthetase with lower affinity for nucleotides. J Biol Chem 1983; 258:4130–4136.

    PubMed  CAS  Google Scholar 

  102. Hsueh CT, Dolnick BJ. Regulation of folate-binding protein gene expression by DNA methylation in methotrexate-resistant KB cells. Biochem Pharmacol 1994; 47:1019–1027.

    Article  PubMed  CAS  Google Scholar 

  103. Cowan KH, Jolivet J. Methotrexate-resistant human breast cancer cell line with multiple defects, including diminished formation of methotrexate polyglutamates. J Biol Chem 1984; 259:10793–10800.

    PubMed  CAS  Google Scholar 

  104. Grant SC, Kris MG, Young CW et al. Edatrexate, an antifolate with antitumor activity: A review. [Review]. Cancer Investg 1993; 11:36–45.

    Article  PubMed  CAS  Google Scholar 

  105. Sirotnak FM, Moccio DM, Kelleher LE et al. Relative frequency and kinetic properties of transport-defective phenotypes among methotrexate-resistant L1210 clonal cell lines derived in vivo. Cancer Res 1981; 41:4447–4452.

    PubMed  CAS  Google Scholar 

  106. Dixon KH, Lanpher BC, Chiu J et al. A novel cDNA restores reducted folate carrier activity and methotrexate sensitivity to transport deficient cells. J Biol Chem 1994; 269:17–20.

    PubMed  CAS  Google Scholar 

  107. Chung KN, Saikawa Y, Paik TH et al. Stable transfectants of human MCF-7 breast cancer cells with increased levels of the human folate receptor exhibit an increased sensitivity to antifolates. J Clin Invest 1993; 91:1289–1294.

    Article  PubMed  CAS  Google Scholar 

  108. Kool M, van der LM, de Haas M et al. MRP3, an organic anion transporter able to transport anti-cancer drugs. Proc Natl Acad Sci USA 1999; 96:6914–6919.

    Article  PubMed  CAS  Google Scholar 

  109. Hooijberg JH, Broxterman HJ, Scheffer GL et al. Potent interaction of flavopiridol with MRP1. British Journal of Cancer 1999; 81:269–276.

    Article  PubMed  CAS  Google Scholar 

  110. Cowan KH, Jolivet J. A methotrexate-resistant human breast cancer cell line with multiple defects, including diminished formation of methotrexate polyglutamates. J Biol Chem 1984; 259:10793–10800.

    PubMed  CAS  Google Scholar 

  111. Volk EL, Rohde K, Rhee M et al. Methotrexate cross-resistance in a mitoxantrone-selected multidrug-resistant MCF7 breast cancer cell line is attributable to enhanced energy-dependent drug efflux. Cancer Res 2000; 60:3514–3521.

    PubMed  CAS  Google Scholar 

  112. Rhee MS, Wang Y, Nair MG et al. Acquisition of resistance to antifolates caused by enhanced gamma-glutamyl hydrolase activity. Cancer Res 1993; 53:2227–2230.

    PubMed  CAS  Google Scholar 

  113. Spears CP. Clinical resistance to antimetabolites. Hematol Oncol Clin North Am 1995; 9:397–413.

    PubMed  CAS  Google Scholar 

  114. Ackland SP, Schilsky RL. High-dose methotrexate: A critical reappraisal. J Clin Oncol 1987; 5:2017–2031.

    PubMed  CAS  Google Scholar 

  115. Kamen BA, Winick NJ. High dose methotrexate therapy: Insecure rationale? Biochem Pharmacol 1988; 37:2713–2715.

    Article  PubMed  CAS  Google Scholar 

  116. Jackson RC, Jackman AL, Calvert AH. Biochemical effects of a quinazoline inhibitor of thymidylate synthetase, N-(4-(N-((2-amino-4-hydroxy-6-quinazolinyl)methyl)prop-2-ynylamino) benzoyl)-L-glutamic acid (CB3717), on human lymphoblastoid cells. Biochem Pharmacol 1983; 32:3783–3790.

    Article  PubMed  CAS  Google Scholar 

  117. Beardsley GP, Moroson BA, Taylor EC et al. A new folate antimetabolite, 5,10-dideaza-5, 6,7,8-tetrahydrofolate is a potent inhibitor of de novo purine synthesis. J Biol Chem 1989; 264;328–333.

    PubMed  CAS  Google Scholar 

  118. Elias L, Crissman HA. Interferon effects upon the adenocarcinoma 38 and HL-60 cell lines: Antiproliferative reseponses and synergistic interactions with halogenated pyrimidine antimetabolites. Cancer Res 1988; 48:4868–4873,.

    PubMed  CAS  Google Scholar 

  119. Auerbach M, Elias EG, Orford J. Experience with methotrexate, 5-fluorouracil, and leucovorin (MFL): A first line effective, minimally toxic regimen for metastatic breast cancer. Cancer Invest 2002; 20:24–28.

    Article  PubMed  CAS  Google Scholar 

  120. Klatt O, Stehlin Jr JS, McBride C et al. The effect of nitrogen mustard treatment on the deoxyribonucleic acid of sensitive and resistant Ehrlich tumor cells. Cancer Res 1969; 29:286–290.

    PubMed  CAS  Google Scholar 

  121. Nogae I, Kohno K, Kikuchi J et al. Analysis of structural features of dihydropyridine analogs needed to reverse mutidrug resistance and to inhibit photoaffinity labelin of P-glycoprotein. Biochem Pharmacol 1989; 38:519–527.

    Article  PubMed  CAS  Google Scholar 

  122. Zamble DB, Lippard SJ. Cisplatin and DNA repair in cancer chemotherapy. Trends Biochem Sci 1995; 20:435–439.

    Article  PubMed  CAS  Google Scholar 

  123. Fairchild CR, Moscow JA, O’Brien EE et al. Multidrug resistance in cells transfected with human genes encoding a variant P-glycoprotein and glutathione S-transferase-pi. Mol Pharmacol 1990; 37:801–809.

    PubMed  CAS  Google Scholar 

  124. Townsend AJ, Tu CP, Cowan KH. Expression of human mu or alpha class glutathione S-transferases in stably transfected human MCF-7 breast cancer cells: Effect on ellular sensitivity to cytotoxic agents. Mol Pharmacol 1992; 41:230–236.

    PubMed  CAS  Google Scholar 

  125. Hilton J. Deoxyribonucleic acid crosslinking by 4-hydroperoxycyclophosphamide in cyclophosphamide-sensitive and-Resistant L1210 cells. Biochem Pharmacol 1984; 33:1867–1872.

    Article  PubMed  CAS  Google Scholar 

  126. Perez RP. Cellular and molecular determinants of cisplatin resistance. Eur J Cancer 1998; 34:1535–1542.

    Article  PubMed  CAS  Google Scholar 

  127. Perez RP. Cellular and molecular determinants of cisplatin resistance. Eur J Cancer 1998; 34:1535–1542.

    Article  PubMed  CAS  Google Scholar 

  128. Mackey JR, Mani RS, Selner M et al. Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res 1998; 58:4349–4357.

    PubMed  CAS  Google Scholar 

  129. Mackey JR, Mani RS, Selner M et al. Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res 1998; 58:4349–4357.

    PubMed  CAS  Google Scholar 

  130. Ruiz VHV, Veerman G, Eriksson S et al. Development and molecular characterization of a 2′,2′-difluorodeoxycytidine-resistant variant of the human ovarian carcinoma cell line A2780. Cancer Res 1994; 54:4138–4143.

    Google Scholar 

  131. Goan YG, Zhou B, Hu E et al. Overexpression of ribonucleotide reductase as a mechanism of resistance to 2,2-difluorodeoxycytidine in the human KB cancer cell line. Cancer Res 1999; 59:4204–4207.

    PubMed  CAS  Google Scholar 

  132. Bergman AM, Pinedo HM Jongsma AP et al. Decreased resistance to gemcitabine (2′,2′-difluorodeoxycitidine) of cytosine arabinoside-resistant myeloblastic murine and rat leukemia cell lines: Role of altered activity and substrate specificity of deoxycytidine kinase. Biochem Pharmacol 1999; 57:397–406.

    Article  PubMed  CAS  Google Scholar 

  133. Schirmer M, Stegmann AP, Geisen F et al. Lack of cross-resistance with gemcitabine and cytarabine in cladribine-resistant HL60 cells with elevated 5′-nucleotidase activity. Exp Hematol 1998; 26:1223–1228.

    PubMed  CAS  Google Scholar 

  134. Abburzzese JL, Grunewald R, Weeks EA et al. A phase I clinical, plasma, and cellular pharmacology study of gemcitabine. J Clin Oncol 1991; 9:491–498.

    Google Scholar 

  135. Roodi N, Bailey LR, Kao WY et al. Estrogen receptor gene analysis in estrogen receptor-positive and receptor-negative primary breast cancer. J Natl Cancer Inst 1995; 87:446–451.

    Article  PubMed  CAS  Google Scholar 

  136. Gottardis MM, Jordan VC. Development of tamoxifen-stimulated growth of MCF-7 tumors in athymic mice after long-term antiestrogen administration. Cancer Res 1988; 48:5183–5187.

    PubMed  CAS  Google Scholar 

  137. Pietras RJ, Arboleda J, Reese DM et al. Her-2 tyrosine kinase pathway targets estrogen receptor and promotes hormone-independent growth in human breast cancer cells. Oncogene 1995; 10:2435–2446.

    PubMed  CAS  Google Scholar 

  138. Gottardis MM, Martin MK, Jordan VC. Long-term tamoxifen therapy to control transplanted human breast tumor growth in athymic mice. In: Salmon SE, ed. Adjuvant Therapy of Cancer V. Orlando: 1987:447–453.

    Google Scholar 

  139. Lavinsky RM, Jepsen K, Heinzel T et al. Diverse signaling pathways modulate nuclear receptor recruitment of N-CoR and SMRT complexes. Proc Natl Acad Sci USA 1998; 95:2920–2925.

    Article  PubMed  CAS  Google Scholar 

  140. Dauvois S, White R, Parker MG. The antiestrogen ICI 182780 disrupts estrogen receptor nucleocytoplasmic shuttling. J Cell Sci 1993; 106(Pt 4):1377–1388.

    PubMed  CAS  Google Scholar 

  141. Parker MG, Arbuckle N, Dauvois S et al. Structure and function of the estrogen receptor. Ann NY Acad Sci 1993; 684:119–126.

    Article  PubMed  CAS  Google Scholar 

  142. Basclga J, Norton L, Albanell J et al. Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Research 1998; 58:2825–2831.

    Google Scholar 

  143. Sliwkowski MX, Lofgren JA, Lewis GD et al. Nonclinical studies addressing the mechanism of action of trastuzumab (Herceptin). Semin Oncol 1999; 26:60–70.

    PubMed  CAS  Google Scholar 

  144. Lewis GD, Figari I, Fendly B et al. Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol Immunother 1993; 37:255–263.

    Article  PubMed  CAS  Google Scholar 

  145. Pegram MD, Baly D, Wirth C et al. Antibody dependent cell-mediated cytotoxicity in breast cancer patients in Phase III clinical trials of a humanized anti-HER2 antibody. Proceedings of the American Association for Cancer Research 1997; 38:602.

    Google Scholar 

  146. Pegram M, Hsu S, Lewis G et al. Inhibitory effects of combinations of HER-2/neu antibody and chemotherapeutic agents used for treatment of human breast cancers. Oncogene 1999; 18:2241–2251.

    Article  PubMed  CAS  Google Scholar 

  147. Argiris A, Wang CX, Whalen SG et al. Synergistic interactions between tamoxifen and trastuzumab (Herceptin). Clin Cancer Res 2004; 10:1409–1420.

    Article  PubMed  CAS  Google Scholar 

  148. Sachdev D, Yee D. The IGF system and breast cancer. [11] Journal Name 2001; 8:197–209.

    CAS  Google Scholar 

  149. Miller KD. The role of ErbB inhibitors in trastuzumab resistance. Oncologist 2004; 9(Suppl 3):16–19.

    Article  PubMed  CAS  Google Scholar 

  150. Moulder SL, Yakes FM, Muthuswamy SK et al. Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res 2001; 61:8887–8895.

    PubMed  CAS  Google Scholar 

  151. Esteva FJ. Monoclonal antibodies, small molecules, and vaccines in the treatment of breast cancer. Oncologist 2004; 9(Suppl 3):4–9.

    Article  PubMed  CAS  Google Scholar 

  152. Chernicky CL, Tan H, Yi L et al. Treatment of murine breast cancer cells with antisense RNA to the type I insulin-like growth factor receptor decreases the level of plasminogen activator transcripts, inhibits cell growth in vitro, and reduces tumorigenesis in vivo. Mol Pathol 2002; 55:102–109.

    Article  PubMed  CAS  Google Scholar 

  153. Lu YH, Zi XL, Zhao YA et al. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). J Natl Cancer Inst 2001; 93:1852–1857.

    Article  PubMed  CAS  Google Scholar 

  154. Nagata Y, Lan KH, Zhou X et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell 2004; 6:117–127.

    Article  PubMed  CAS  Google Scholar 

  155. Schrag D, Garewal HS, Burstein HJ et al. American society of clinical oncology technology assessment: Chemotherapy sensitivity and resistance assays. J Clin Oncol 2004; 22:3631–3638.

    Article  PubMed  CAS  Google Scholar 

  156. Xu JM, Song ST, Tang ZM et al. Predictive chemotherapy of advanced breast cancer directed by MTT assay in vitro. Breast Cancer Res Treat 1999; 53:77–85.

    Article  PubMed  CAS  Google Scholar 

  157. Symmans WF, Ayers M, Clark EA et al. Total RNA yield and microarray gene expression profiles from fine-needle aspiration biopsy and coreneedle biopsy samples of breast carcinoma. Cancer 2003; 97:2960–71.

    Article  PubMed  CAS  Google Scholar 

  158. Chang JC, Wooten EC, Tsimelzon A et al. Gene expression profiling for the prediction of therapeutic response to docetaxel in patients with breast cancer. Lancet 2003; 2(362):362–9.

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gabriel N. Hortobagyi .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2007 Landes Bioscience and Springer Science+Business Media

About this chapter

Cite this chapter

Gonzalez-Angulo, A.M., Morales-Vasquez, F., Hortobagyi, G.N. (2007). Overview of Resistance to Systemic Therapy in Patients with Breast Cancer. In: Yu, D., Hung, MC. (eds) Breast Cancer Chemosensitivity. Advances in Experimental Medicine and Biology, vol 608. Springer, New York, NY. https://doi.org/10.1007/978-0-387-74039-3_1

Download citation

Publish with us

Policies and ethics