Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-jr42d Total loading time: 0 Render date: 2024-04-23T09:03:15.049Z Has data issue: false hasContentIssue false

41 - Prostate Cancer Metastasis: Thoughts on Biology and Therapeutics

from PART II - CLINICAL RESEARCH

Published online by Cambridge University Press:  05 June 2012

Kosuke Mizutani
Affiliation:
University of Michigan Comprehensive Cancer Center, United States
Russell S. Taichman
Affiliation:
The University of Chicago, Committee on Cancer Biology and Pritzker School of Medicine, United States
Kenneth J. Pienta
Affiliation:
University of Michigan Comprehensive Cancer Center, United States
David Lyden
Affiliation:
Weill Cornell Medical College, New York
Danny R. Welch
Affiliation:
Weill Cornell Medical College, New York
Bethan Psaila
Affiliation:
Imperial College of Medicine, London
Get access

Summary

Prostate cancer accounts for 25 percent of all cancer diagnoses in American men (186,320 in 2008) as well as 10 percent of cancer deaths in men (28,660 in 2008) [1]. Although mortality has fallen by 25 percent over the past decade and the five-year survival is approaching 100 percent, several challenges to decreasing the morbidity and mortality from metastatic prostate cancer remain.

The end result of the process of metastasis in prostate cancer is clear; nearly 100 percent of men suffer from osteoblastic bone metastases at the time of death (Figure 41.1) [2–5]. The mechanisms by which prostate cancer metastasizes to bone sites remain obscure, but there is no doubt that forces that drive preferential seeding are at play. In 1889, Stephen Paget proposed the “seed-and-soil” theory to explain the nonrandom pattern of cancer metastasis [6]. His theory suggested that factors within the metastatic site promote growth, which is analogous to the tendency of seeds to grow in fertile soil (i.e., factors in the environment of the metastatic site could contribute to the proliferation of cancer cells). In 1928, James Ewing proposed that cancer cells grow at a particular site because they are directed to that site by the direction of blood flow and lymphatics [7]. It appears that both these theories have correct elements. Isaiah Fidler defined the modern “seed-and-soil” hypothesis as consisting of three principles [8]. First, cancerous tissues contain heterogeneous subpopulations of cells with different angiogenic, invasive, and metastatic properties.

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 456 - 464
Publisher: Cambridge University Press
Print publication year: 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

,American Cancer Society, http://www.cancer.org/docroot/stt/stt_0.asp?from=fast.
Kingsley, , Fournier, PG, Chirgwin, JM, Guise, TA (2007) Molecular biology of bone metastasis. Mol Cancer Ther. 6(10): 2609–17.CrossRefGoogle ScholarPubMed
Keller, ET, Dai, J, Escara-Wilke, J, Hall, CL et al. (2007) New trends in the treatment of bone metastasis. J Cell Biochem. 102(5): 1095–102.CrossRefGoogle ScholarPubMed
Loberg, RD, Logothetis, CJ, Keller, ET, Pienta, KJ (2005) Pathogenesis and treatment of prostate cancer bone metastases: targeting the lethal phenotype. J Clin Oncol. 23(32): 8232–41.CrossRefGoogle ScholarPubMed
,Washington University (St. Louis) School of Medicine, http://gamma.wustl.edu/bs045te143.html.
Paget, S (1889)The distribution of secondary growths in cancer of the breast. Lancet. 1: 571–3.CrossRefGoogle Scholar
Ewing, J (1928) Metastasis. In Neoplastics (3d ed.). Saunders, Philadelphia, pp. 77–89.Google Scholar
Fidler, IJ (2003) The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 3: 453–8.CrossRefGoogle ScholarPubMed
Pienta, KJ, Loberg, R (2005) The “emigration, migration, and immigration” of prostate cancer. Clin Prostate Cancer. 4(1): 24–30.CrossRefGoogle ScholarPubMed
Sugarbaker, PH (1993) Metastatic inefficiency: The scientific basis for resection of liver metastases from colorectal cancer. J Surg Oncol. 3 (Suppl): 158–60.CrossRefGoogle ScholarPubMed
Kalikin, LM, Schneider, A, Thakur, MA et al. (2003) In vivo visualization of metastatic prostate cancer and quantitation of disease progression in immunocompromised mice. Cancer Biol Ther. 2: 656–60.CrossRefGoogle ScholarPubMed
Rosol, TJ, Tannehill-Gregg, SH, LeRoy, BE, Mandl, S, Contag, CH (2003) Animal models of bone metastasis. Cancer. 97(Suppl): 748–57.CrossRefGoogle ScholarPubMed
Shah, RB, Mehra, R, Chinnaiyan, AM et al. (2004) Androgen-independent prostate cancer is a heterogeneous group of diseases: lessons from a rapid autopsy program. Cancer Res. 64(24): 9209–16.CrossRefGoogle ScholarPubMed
Weiss, L (1988) Biomechanical destruction of cancer cells in the heart: a rate regulator for hematogenous metastasis. Invasion Metastasis. 8: 228–37.Google ScholarPubMed
Weiss, L, Harlos, JP, Elkin, G (1989) Mechanism of mechanical trauma to Ehrlich ascites tumor cells in vitro and its relationship to rapid intravascular death during metastasis. Int J Cancer. 44: 143–8.CrossRefGoogle ScholarPubMed
Shiozawa, Y, Havens, AM, Pienta, KJ, Taichman, RS (2008) The bone marrow niche: habitat to hematopoietic and mesenchymal stem cells, and unwitting host to molecular parasites. Leukemia. 22: 941–50.CrossRefGoogle ScholarPubMed
Taichman, RS, Loberg, RD, Mehra, R, Pienta, KJ (2007) The evolving biology and treatment of prostate cancer. J Clin Invest. 117(9): 2351–61.CrossRefGoogle ScholarPubMed
Taichman, RS, Cooper, C, Keller, ET, Pienta, KJ, Taichman, N, McCauley, LK (2002) Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 62: 1832–7.Google ScholarPubMed
Sun, YX, Wang, J, Shelburne, CE et al. (2003) The expression of CXCR4 and CXCL12 (SDF-1) in human prostate cancers (PCa) in vivo. J Cell Biochem. 89: 462–73.CrossRefGoogle ScholarPubMed
Heresi, GA, Wang, J, Taichman, R et al. (2005) Expression of the chemokine receptor CCR7 in prostate cancer presenting with generalized lymphadenopathy: Report of a case, review of the literature, and analysis of chemokine receptor expression. Urol Oncol Sem Orig Invest. 23: 261–7.Google ScholarPubMed
Liang, ZX, Wu, T, Lou, H et al. (2004) Inhibition of breast cancer metastasis by selective synthetic polypeptide against CXCR4. Cancer Res. 64: 4302–8.CrossRefGoogle ScholarPubMed
Wang, JH, Wang, J, Sun, Y-X et al. (2005) Diverse signaling pathways through the SDF-1/CXCR4 chemokine axis in prostate cancer cell lines leads to altered patterns of cytokine secretion and angiogenesis. Cellular Signalling. 17: 1578–92.CrossRefGoogle ScholarPubMed
Sun, YX, Schneider, A, Jung, Y et al. (2005) Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. J Bone Miner Res. 20: 318–29.CrossRefGoogle ScholarPubMed
Wang, J, Wang, J, Dai, J et al. (2007) A glycolytic mechanism regulating an angiogenic switch in prostate cancer. Cancer Res. 67: 149–59.CrossRefGoogle ScholarPubMed
Wang, J, Loberg, R, Taichman, RS (2006) The pivotal role of CXCL12 (SDF-1)/CXCR4 axis in bone metastasis. Cancer Metastasis Rev. 25: 573–87.CrossRefGoogle ScholarPubMed
Jung, Y, Wang, J, Song, J et al. (2007) Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation. Blood. 110(1): 82–90.CrossRefGoogle ScholarPubMed
Zhu, J, Garrett, R, Jung, Y et al. (2007) Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood. 109(9): 3706–12.CrossRefGoogle ScholarPubMed
Avecilla, ST, Hattori, K, Heissig, B et al. (2004) Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nature Med. 10: 64–71.CrossRefGoogle ScholarPubMed
Tuszynski, GP, Sharma, M., Rothman, VL, Sharma, MC (2002) Angiostatin binds to tyrosine kinase substrate annexin II through the lysine-binding domain in endothelial cells. Microvasc Res. 64: 448–62.CrossRefGoogle ScholarPubMed
Imbriaco, M, Larson, SM, Yeung, HW et al. (1998) A new parameter for measuring metastatic bone involvement by prostate cancer: the Bone Scan Index. Clin Cancer Res. 4(7): 1765–72.Google ScholarPubMed
Hricak, H, Choyke, PL, Eberhardt, SC, Leibel, SA, Scardino, PT (2007) Imaging prostate cancer: a multidisciplinary perspective. Radiology. 243(1): 28–53.CrossRefGoogle ScholarPubMed
Akin, O, Hricak, H (2007) Imaging of prostate cancer. Radiol Clin North Am. 45(1):207–22.CrossRefGoogle ScholarPubMed
Kundra, V (2006) Prostate cancer imaging. Semin Roentgenol. 41(2): 139–49.CrossRefGoogle ScholarPubMed
Even-Sapir, E, Metser, U, Mishani, E, Lievshitz, G, Lerman, H, Leibovitch, I (2006) The detection of bone metastases in patients with high-risk prostate cancer: 99mTc-MDP Planar bone scintigraphy, single- and multi-field-of-view SPECT, 18F-fluoride PET, and 18F-fluoride PET/CT. J Nucl Med. 47(2): 287–97.Google ScholarPubMed
Husarik, DB, Miralbell, R, Dubs, M et al. (2008) Evaluation of [(18)F]-choline PET/CT for staging and restaging of prostate cancer. Eur J Nucl Med Mol Imaging. 35(2): 253–63.CrossRefGoogle ScholarPubMed
Lecouvet, FE, Geukens, D, Stainier, A et al. (2007) Magnetic resonance imaging of the axial skeleton for detecting bone metastases in patients with high-risk prostate cancer: diagnostic and cost-effectiveness and comparison with current detection strategies. J Clin Oncol. 25(22): 3281–7.CrossRefGoogle ScholarPubMed
Nakanishi, K, Kobayashi, M, Nakaguchi, K et al. (2007) Whole-body MRI for detecting metastatic bone tumor: diagnostic value of diffusion-weighted images. Magn Reson Med Sci. 6(3):147–55CrossRefGoogle ScholarPubMed
Schmidt, GP, Schoenberg, SO, Schmid, R et al. (2007) Screening for bone metastases: whole-body MRI using a 32-channel system versus dual-modality PET-CT. Eur Radiol. 17(4): 939–49.CrossRefGoogle ScholarPubMed
Therasse, P, Arbuck, SG, Eisenhauer, EA et al. (2000) New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 92(3): 205–16.CrossRefGoogle ScholarPubMed
Scher, HI, Halabi, S, Tannock, I et al. Prostate Cancer Clinical Trials Working Group. (2008) Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 26(7): 1148–59.CrossRefGoogle ScholarPubMed
Lee, KC, Bradley, DA, Hussain, M et al. (2007) A feasibility study evaluating the functional diffusion map as a predictive imaging biomarker for detection of treatment response in a patient with metastatic prostate cancer to the bone. Neoplasia. 9(12): 1003–11.CrossRefGoogle Scholar
Koopmans, N, Jong, IJ, Breeuwsma, AJ, Veer, E (2007) Serum bone turnover markers (PINP and ICTP) for the early detection of bone metastases in patients with prostate cancer: a longitudinal approach. J Urol. 178(3 Pt 1): 849–53.CrossRefGoogle ScholarPubMed
Jung, K, Lein, M, Stephan, C et al. (2004) Comparison of 10 serum bone turnover markers in prostate carcinoma patients with bone metastatic spread: diagnostic and prognostic implications. Int J Cancer. 111(5): 783–91.CrossRefGoogle ScholarPubMed
Hegele, A, Wahl, HG, Varga, Z et al. (2007) Biochemical markers of bone turnover in patients with localized and metastasized prostate cancer. BJU Int. 99(2): 330–4.CrossRefGoogle ScholarPubMed
Garnero, P, Buchs, N, Zekri, J, Rizzoli, R, Coleman, RE, Delmas, PD (2000) Markers of bone turnover for the management of patients with bone metastases from prostate cancer. Br J Cancer. 82(4): 858–64.CrossRefGoogle ScholarPubMed
Lein, M, Wirth, M, Miller, K et al. (2007) Serial markers of bone turnover in men with metastatic prostate cancer treated with zoledronic acid for detection of bone metastases progression. Eur Urol. 52(5): 1381–7.CrossRefGoogle ScholarPubMed
Clines, GA, Guise, TA (2008) Molecular mechanisms and treatment of bone metastasis. Expert Rev Mol Med. 10: e7.CrossRefGoogle ScholarPubMed
Clezardin, P, Teti, A (2007) Bone metastasis: pathogenesis and therapeutic implications. Clin Exp Metastasis. 24(8): 599–608.CrossRefGoogle ScholarPubMed
Lipton, A (2006) Future treatment of bone metastases. Clin Cancer Res. 12(20 Pt 2): 6305s–6308s.CrossRefGoogle ScholarPubMed
Pienta, KJ, Smith, DC (2005) Advances in prostate cancer chemotherapy: a new era begins. CA Cancer J Clin. 55: 300–18.CrossRefGoogle ScholarPubMed
Scher, HI, Sawyers, CL (2005) Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol. 23: 8253–61.CrossRefGoogle ScholarPubMed
Pienta, KJ, Bradley, D (2006) Mechanisms underlying the development of androgen-independent prostate cancer. Clin Cancer Res. 12(6):1665–71.CrossRefGoogle ScholarPubMed
Huggins, CB, Hodges, CV (1941) Studies on prostatic cancer. 1. The effect of castration, of estrogen and of androgen injections on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1: 293–7.Google Scholar
Huggins, C (1967) Endocrine-induced regression of cancers. Cancer Res. 27: 1925–30.Google ScholarPubMed
Tannock, IF, Wit, R, Berry, WR et al. (2004) Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 351: 1502–12.CrossRefGoogle ScholarPubMed
Garmey, EG, Sartor, O, Halabi, S, Vogelzang, NJ (2008) Second-line chemotherapy for advanced hormone-refractory prostate cancer. Clin Adv Hematol Oncol. 6(2): 118–32.Google ScholarPubMed
Bradley, NM, Husted, J, Sey, MS et al. (2008) Did the pattern of practice in the prescription of palliative radiotherapy for the treatment of uncomplicated bone metastases change between 1999 and 2005 at the Rapid Response Radiotherapy Program? Clin Oncol (R Coll Radiol) 20: 327–36.CrossRefGoogle ScholarPubMed
Venkitaraman, R, Barbachano, Y, Dearnaley, DP et al. (2007) Outcome of early detection and radiotherapy for occult spinal cord compression. Radiother Oncol. 85(3): 469–72.CrossRefGoogle ScholarPubMed
Wu, JS, Wong, R, Johnston, M, Bezjak, A, Whelan, T. Cancer Care Ontario Practice Guidelines Initiative Supportive Care Group. (2003) Meta-analysis of dose-fractionation radiotherapy trials for the palliation of painful bone metastases. Int J Radiat Oncol Biol Phys. 55(3): 594–605.CrossRefGoogle Scholar
Baczyk, M, Czepczyński, R, Milecki, P, Pisarek, M, Oleksa, R, Sowiński, J (2007) 89Sr versus 153Sm-EDTMP: comparison of treatment efficacy of painful bone metastases in prostate and breast carcinoma. Nucl Med Commun. 28(4): 245–50.CrossRefGoogle ScholarPubMed
Dolezal, J, Vizda, J, Odrazka, K (2007) Prospective evaluation of samarium-153-EDTMP radionuclide treatment for bone metastases in patients with hormone-refractory prostate cancer. Urol Int. 78(1): 50–7.CrossRefGoogle ScholarPubMed
Liepe, K, Kotzerke, J (2007) A comparative study of 188Re-HEDP, 186Re-HEDP, 153Sm-EDTMP and 89Sr in the treatment of painful skeletal metastases. Nucl Med Commun. 28(8): 623–30.CrossRefGoogle ScholarPubMed
Greenspan, SL (2008) Approach to the prostate cancer patient with bone disease. J Clin Endocrinol Metab. 93(1): 2–7.CrossRefGoogle ScholarPubMed
Dhillon, S, Lyseng-Williamson, KA (2008) Zoledronic acid: a review of its use in the management of bone metastases of malignancy. Drugs. 68(4): 507–34.CrossRefGoogle Scholar
McKeage, K, Plosker, GL (2008) Zoledronic acid: a pharmacoeconomic review of its use in the management of bone metastases. Pharmacoeconomics. 26(3): 251–68.CrossRefGoogle ScholarPubMed
Saad, F (2008) New research findings on zoledronic acid: survival, pain, and anti-tumour effects. Cancer Treat Rev. 34(2): 183–92.CrossRefGoogle ScholarPubMed
Hamdy, N (2008) Denosumab: RANKL inhibition in the management of bone loss. Drugs Today (Barc). 44(1): 7–21.CrossRefGoogle ScholarPubMed
Vega, D, Maalouf, NM, Sakhaee, K (2007) The role of receptor activator of nuclear factor-kappaB (RANK)/RANK ligand/osteoprotegerin: clinical implications. J Clin Endocrinol Metab. 92(12): 4514–21.CrossRefGoogle ScholarPubMed
Kearns, AE, Khosla, S, Kostenuik, P (2007) RANKL and OPG regulation of bone remodeling in health and disease. Endocr Rev. 29: 155–92.CrossRefGoogle ScholarPubMed
Lalich, M, McNeel, DG, Wilding, G, Liu, G (2007) Endothelin receptor antagonists in cancer therapy. Cancer Invest. 25(8): 785–94.CrossRefGoogle ScholarPubMed
Carducci, MA, Jimeno, A (2006) Targeting bone metastasis in prostate cancer with endothelin receptor antagonists. Clin Cancer Res. 12(20 Pt 2): 6296s–6300s.CrossRefGoogle ScholarPubMed
Carducci, MA, Saad, F, Abrahamsson, PA et al. Atrasentan Phase III Study Group Institutions. (2007) A Phase 3 randomized controlled trial of the efficacy and safety of atrasentan in men with metastatic hormone-refractory prostate cancer. Cancer. 110(9): 1959–66.CrossRefGoogle Scholar
Logothetis, CJ, Navone, NM, Lin, SH (2008) Understanding the biology of bone metastases: key to the effective treatment of prostate cancer. Clin Cancer Res. 14(6): 1599–602.CrossRefGoogle ScholarPubMed
Tu, SM, Lin, SH (2008) Current trials using bone-targeting agents in prostate cancer. Cancer J. 14(1): 5–9.CrossRefGoogle ScholarPubMed
Aragon-Ching, JB, Dahut, WL (2008) The role of angiogenesis inhibitors in prostate cancer. Cancer J. 14(1): 20–5.CrossRefGoogle ScholarPubMed
Harzstark, AL, Ryan, CJ (2008) Novel therapeutic strategies in development for prostate cancer. Expert Opin Investig Drugs. 17(1): 13–22.CrossRefGoogle ScholarPubMed
Di Lorenzo, G, Figg, WD, Fossa, SD et al. (2008) Combination of bevacizumab and docetaxel in docetaxel-pretreated hormone-refractory prostate cancer: a Phase 2 study. Eur Urol. 54: 1089–94.CrossRefGoogle ScholarPubMed
Efstathiou, E, Troncoso, P, Wen, S et al. (2007) Initial modulation of the tumor microenvironment accounts for thalidomide activity in prostate cancer. Clin Cancer Res. 13(4): 1224–31.CrossRefGoogle ScholarPubMed
Aragon-Ching, JB, Li, H, Gardner, ER, Figg, WD (2007) Thalidomide analogues as anticancer drugs. Recent Patents Anticancer Drug Discov. 2(2): 167–74.CrossRefGoogle ScholarPubMed
Ryan, CJ, Stadler, WM, Roth, B et al. (2007) Phase I dose escalation and pharmacokinetic study of AZD2171, an inhibitor of the vascular endothelial growth factor receptor tyrosine kinase, in patients with hormone refractory prostate cancer (HRPC). Invest New Drugs. 25(5): 445–51.CrossRefGoogle ScholarPubMed
Beekman, KW, Colevas, AD, Cooney, K et al. (2006) Phase II evaluations of cilengitide in asymptomatic patients with androgen-independent prostate cancer: scientific rationale and study design. Clin Genitourin Cancer. 4(4): 299–302.CrossRefGoogle ScholarPubMed
Mullamitha, SA, Ton, NC, Parker, GJ et al. (2007) Phase I evaluation of a fully human anti-alphaV integrin monoclonal antibody (CNTO 95) in patients with advanced solid tumors. Clin Cancer Res. 13(7): 2128–35.CrossRefGoogle ScholarPubMed
Gramoun, A, Shorey, S, Bashutski, JD et al. (2007) Effects of vitaxin, a novel therapeutic in trial for metastatic bone tumors, on osteoclast functions in vitro. J Cell Biochem. 102(2): 341–52.CrossRefGoogle ScholarPubMed
Ward, JE, McNeel, DG (2007) GVAX: an allogeneic, whole-cell, GM-CSF-secreting cellular immunotherapy for the treatment of prostate cancer. Expert Opin Biol Ther. 7(12): 1893–902.CrossRefGoogle ScholarPubMed
Doehn, C, Böhmer, T, Kausch, I, Sommerauer, M, Jocham, D (2008) Prostate cancer vaccines: current status and future potential. BioDrugs. 22(2): 71–84.CrossRefGoogle ScholarPubMed
Patel, PH, Kockler, DR (2008) Sipuleucel-T: a vaccine for metastatic, asymptomatic, androgen-independent prostate cancer. Ann Pharmacother. 42(1): 91–8.CrossRefGoogle ScholarPubMed
Noonan, DM, Lerma Barbaro, A, Vannini, N, Mortara, L, Albini, A (2008) Inflammation, inflammatory cells and angiogenesis: decisions and indecisions. Cancer Metastasis Rev. 27(1): 31–40.CrossRefGoogle ScholarPubMed
Allavena, P, Sica, A, Solinas, G, Porta, C, Mantovani, A (2008) The inflammatory micro-environment in tumor progression: The role of tumor-associated macrophages. Crit Rev Oncol Hematol. 66(1): 1–9.CrossRefGoogle ScholarPubMed
Porta, C, Subhra Kumar, B, Larghi, P, Rubino, L, Mancino, A, Sica, A (2007) Tumor promotion by tumor-associated macrophages. Adv Exp Med Biol. 604: 67–86.CrossRefGoogle ScholarPubMed
Sica, A, Schioppa, T, Mantovani, A, Allavena, P (2006) Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer. 42(6): 717–27.CrossRefGoogle ScholarPubMed
Loberg, RD, Ying, C, Craig, M et al. (2007) Targeting CCL2 with systemic delivery of neutralizing antibodies induces prostate cancer tumor regression in vivo. Cancer Res. 67(19): 9417–24.CrossRefGoogle ScholarPubMed
Loberg, RD, Ying, C, Craig, M et al. (2007) CCL2 as an important mediator of prostate cancer growth in vivo through the regulation of macrophage infiltration. Neoplasia. 9(7): 556–62.CrossRefGoogle ScholarPubMed
Wallner, L, Dai, J, Escara-Wilke, J et al. (2006) Inhibition of interleukin-6 with CNTO328, an anti-interleukin-6 monoclonal antibody, inhibits conversion of androgen-dependent prostate cancer to an androgen-independent phenotype in orchiectomized mice. Cancer Res. 66(6): 3087–95.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×