Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-m42fx Total loading time: 0 Render date: 2024-07-16T21:13:14.726Z Has data issue: false hasContentIssue false

7 - Principles of intra-arterial infusional chemotherapy in the treatment of liver metastases from colorectal cancer

from Section II - Principles of image-guided therapies

Published online by Cambridge University Press:  05 September 2016

Fidel David Huitzil Melendez
Affiliation:
Memorial Sloan Kettering Cancer Center
Nancy Kemeny
Affiliation:
Memorial Sloan Kettering Cancer Center
Jean-Francois H. Geschwind
Affiliation:
Yale University School of Medicine, Connecticut
Michael C. Soulen
Affiliation:
Department of Radiology, University of Pennsylvania Hospital, Philadelphia
Get access

Summary

Background

The rationale for regional chemotherapy is to maximize drug concentrations and tumor drug uptake in the target organ and minimize systemic toxicity. For regional drug delivery to successfully impact relevant outcomes, several important principles regarding tumor biology, drug pharmacology, and delivery systems must be fulfilled. The model of liver metastasis from colorectal cancer complies with these principles, as colorectal cancer has a regional pattern of dissemination, with the liver being the only site of metastatic disease for long periods of time in some cases. Other salient features include the selective supply of blood to liver metastases by the hepatic artery and availability of active drugs with suitable pharmacokinetic properties.

In this chapter, pharmacological concepts of regional therapy will be reviewed. Desirable pharmacokinetic and pharmacodynamic characteristics of drugs considered for evaluation in the hepatic arterial infusion (HAI) model of regional drug delivery will be discussed, using floxuridine (FUDR) as a model, in the context of patients with colorectal liver-only metastases (CRLM). Also, an evaluation of the pharmacology of currently approved agents for the treatment of metastatic colon cancer and their hypothetical value for regional drug delivery will be performed. Results of phase I and phase II trials of HAI with these drugs, if available, will be explained in terms of the previously established pharmacologic rationale. Finally, for active drugs not already tested, a critical analysis will be done of the available pharmacokinetics data to determine the likelihood of successful incorporation of these drugs to HAI-based therapeutics. We provide an appendix with basic pharmacology definitions to enhance comprehension for the reader who is not familiar with these concepts.

Pharmacologic concepts useful in understanding and evaluating potential advantages of regional delivery for specific drugs

The ultimate goal of regional therapy is to improve the therapeutic index by increasing efficacy and decreasing systemic toxicity. Hepatic arterial therapy relies on two important assumptions:

  1. 1. Regional delivery of the drug leads to increased local concentration and therefore increased therapeutic response.

  2. 2. Regional delivery of the drug leads to decreased systemic exposure and reduced systemic toxicity.

The suitability of any specific drug for regional therapy can be evaluated by the extent to which it fulfills these assumptions.

Type
Chapter
Information
Interventional Oncology
Principles and Practice of Image-Guided Cancer Therapy
, pp. 52 - 64
Publisher: Cambridge University Press
Print publication year: 2016

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

1. Collins, JM. Pharmacologic rationale for regional drug delivery. J Clin Oncol 1984; 2 (5): 498–504.Google Scholar
2. Kemeny, NE., et al. Intra-arterial chemotherapy for liver tumors. In Blumgart, LH, ed. Surgery of the Liver, Biliary Tract and Pancreas. Philadelphia, PA: Saunders Elsevier, 2007; pp. 1321–1337.
3. Weiss, L, et al. Haematogenous metastatic patterns in colonic carcinoma: an analysis of 1541 necropsies. J Pathol 1986; 150 (3): 195–203.Google Scholar
4. Sigurdson, ER, et al. Tumor and liver drug uptake following hepatic artery and portal vein infusion. J Clin Oncol 1987; 5 (11): 1836–1840.Google Scholar
5. Ensminger, WD, et al. A clinical–pharmacological evaluation of hepatic arterial infusions of 5-fluoro-2′-deoxyuridine and 5-fluorouracil. Cancer Res 1978; 38 (11): 3784–3792.Google Scholar
6. Arteaga, CL, Baselga, J. Clinical trial design and end points for epidermal growth factor receptor-targeted therapies: implications for drug development and practice. Clin Cancer Res 2003; 9 (5): 1579–1589.Google Scholar
7. Ensminger, WD. Intrahepatic arterial infusion of chemotherapy: pharmacologic principles. Semin Oncol 2002; 29 (2): 119–125.Google Scholar
8. Laar, JA Van, et al. Comparison of 5-fluoro-2′-deoxyuridine with 5-fluorouracil and their role in the treatment of colorectal cancer. Eur J Cancer 1998; 34 (3): 296–306.Google Scholar
9. Park, JG, et al. Enhancement of fluorinated pyrimidine-induced cytotoxicity by leucovorin in human colorectal carcinoma cell lines. J Natl Cancer Inst 1988; 80 (19): 1560–1564.Google Scholar
10. Ragnhammar, P, et al. A systematic overview of chemotherapy effects in colorectal cancer. Acta Oncol 2001; 40 (2–3): 282–308.Google Scholar
11. Wagner, JG, et al. Steady-state nonlinear pharmacokinetics of 5-fluorouracil during hepatic arterial and intravenous infusions in cancer patients. Cancer Res 1986; 46 (3): 1499–1506.Google Scholar
12. Boublil, JL, et al. Continuous 5-day regional chemotherapy by 5-fluorouracil in colon carcinoma: pharmacokinetic evaluation. Br J Cancer 1985; 52 (1): 15–20.Google Scholar
13. Goldberg, JA, et al. Pharmacokinetics and pharmacodynamics of locoregional 5 fluorouracil (5FU) in advanced colorectal liver metastases. Br J Cancer 1988; 57 (2): 186–189.Google Scholar
14. Grage, TB, et al. Results of a prospective randomized study of hepatic artery infusion with 5-fluorouracil versus intravenous 5-fluorouracil in patients with hepatic metastases from colorectal cancer: a Central Oncology Group study. Surgery 1979; 86 (4): 550–555.Google Scholar
15. Iyer, L, Ratain, MJ. Clinical pharmacology of camptothecins. Cancer Chemother Pharmacol 1998; 42 Suppl: S31–S43.Google Scholar
16. Abigerges, D, et al. Phase I and pharmacologic studies of the camptothecin analog irinotecan administered every 3 weeks in cancer patients. J Clin Oncol 1995; 13(1): 210–221.Google Scholar
17. Herben, VM, et al. Phase I and pharmacokinetic study of irinotecan administered as a low-dose, continuous intravenous infusion over 14 days in patients with malignant solid tumors. J Clin Oncol 1999; 17 (6): 1897–1905.Google Scholar
18. Ohe, Y, et al. Phase I study and pharmacokinetics of CPT-11 with 5-day continuous infusion. J Natl Cancer Inst 1992; 84 (12): 972–974.Google Scholar
19. Riel, JM Van, et al. Continuous administration of irinotecan by hepatic arterial infusion: a phase I and pharmacokinetic study. Clin Cancer Res 2002; 8 (2): 405–412.Google Scholar
20. Riel, JM Van, et al. Continuous infusion of hepatic arterial irinotecan in pretreated patients with colorectal cancer metastatic to the liver. Ann Oncol 2004; 15 (1): 59–63.Google Scholar
21. Fiorentini, G, et al. Irinotecan hepatic arterial infusion chemotherapy for hepatic metastases from colorectal cancer: results of a phase I clinical study. Tumori 2001; 87 (6): 388–390.Google Scholar
22. Desoize, B, Madoulet, C. Particular aspects of platinum compounds used at present in cancer treatment. Crit Rev Oncol Hematol 2002; 42 (3): 317–325.Google Scholar
23. Kweekel, DM, Gelderblom, H, Guchelaar, HJ. Pharmacology of oxaliplatin and the use of pharmacogenomics to individualize therapy. Cancer Treat Rev 2005; 31 (2): 90–105.Google Scholar
24. Kornmann, M, et al. Oxaliplatin exerts potent in vitro cytotoxicity in colorectal and pancreatic cancer cell lines and liver metastases. Anticancer Res 2000; 20 (5A): 3259–3264.Google Scholar
25. Raymond, E, et al. Oxaliplatin: a review of preclinical and clinical studies. Ann Oncol 1998; 9 (10): 1053–1071.Google Scholar
26. Rothenberg, ML, et al. Superiority of oxaliplatin and fluorouracil-leucovorin compared with either therapy alone in patients with progressive colorectal cancer after irinotecan and fluorouracil–leucovorin: interim results of a phase III trial. J Clin Oncol 2003; 21 (11): 2059–2069.Google Scholar
27. Tournigand, C, et al. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol 2004; 22 (2): 229–237.Google Scholar
28. Graham, MA, et al. Clinical pharmacokinetics of oxaliplatin: a critical review. Clin Cancer Res 2000; 6 (4): 1205–1218.Google Scholar
29. Kern, W, et al. Phase I and pharmacokinetic study of hepatic arterial infusion with oxaliplatin in combination with folinic acid and 5-fluorouracil in patients with hepatic metastases from colorectal cancer. Ann Oncol 2001; 12 (5): 599–603.Google Scholar
30. Guthoff, I, et al. Hepatic artery infusion using oxaliplatin in combination with 5-fluorouracil, folinic acid and mitomycin C: oxaliplatin pharmacokinetics and feasibility. Anticancer Res 2003; 23 (6D): 5203–5208.Google Scholar
31. Mancuso, A, et al. Hepatic arterial continuous infusion (HACI) of oxaliplatin in patients with unresectable liver metastases from colorectal cancer. Anticancer Res 2003; 23 (2C): 1917–1922.Google Scholar
32. Fiorentini, G, et al. Oxaliplatin hepatic arterial infusion chemotherapy for hepatic metastases from colorectal cancer: a phase I–II clinical study. Anticancer Res 2004; 24 (3b): 2093–2096.Google Scholar
33. Ducreux, M, et al. Hepatic arterial oxaliplatin infusion plus intravenous chemotherapy in colorectal cancer with inoperable hepatic metastases: a trial of the gastrointestinal group of the Federation Nationale des Centres de Lutte Contre le Cancer. J Clin Oncol 2005; 23 (22): 4881–4887.Google Scholar
34. Levi, F, Hebbar, M, Smith, D, Lepère, C, Focan, CNJ, et al. Final results of first European phase II trial of intravenous cetuximab (Cet) and hepatic artery infusion of irinotecan, 5-fluorouracil, and oxaliplatin in patients (pts) with unresectable liver metastases from wt KRAS colorectal cancer (LM-CRC) after systemic treatment failure (OPTILIV, NCT00852228). In 2013 Gastrointestina Cancers Symposium. 2013. San Francisco, CA: Journal of Clinical Oncology.
35. Bouchahda, M, Grimaldi, MCE, Chatelut, E, Innominato, PF, Paintaud, G, et al. Chronomodulated hepatic artery infusion (HAI) of irinotecan, 5-fluorouracil (5-FU), and oxaliplatin (l-OHP) plus intravenous (iv) cetuximab (Cet) (Chrono-Optiliv) in patients with unresectable liver metastases from wt KRAS colorectal cancer (LM-CRC) after treatment failure (European phase II trial NCT 00852228). In 2013 ASCO Annual Meeting. 2013. Chicago, IL.
36. Fiorentini, G, et al. Intra-arterial infusion of irinotecan-loaded drug-eluting beads (DEBIRI) versus intravenous therapy (FOLFIRI) for hepatic metastases from colorectal cancer: final results of a phase III study. Anticancer Res 2012; 32 (4): 1387–1395.Google Scholar
37. Salman, HS, Cynamon, J, Jagust, M, et al. Randomized phase II trial of embolization versus chemoembolization therapy in previously treated patients with colorectal carcinoma metastatic to the liver. Clin Colorectal Cancer 2002; 2 (3): 173–179.Google Scholar
38. Chen, X, et al. Quantification of irinotecan, SN38, and SN38G in human and porcine plasma by ultra high-performance liquid chromatography-tandem mass spectrometry and its application to hepatic chemoembolization. J Pharm Biomed Anal 2012; 62: 140–148.Google Scholar
39. Lu, JF, et al. Clinical pharmacokinetics of bevacizumab in patients with solid tumors. Cancer Chemother Pharmacol 2008; 62 (5): 779–786.Google Scholar
40. Chung, KY, et al. Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry. J Clin Oncol 2005; 23 (9): 1803–1810.Google Scholar
41. Scaltriti, M, Baselga, J. The epidermal growth factor receptor pathway: a model for targeted therapy. Clin Cancer Res 2006; 12 (18): 5268–5272.Google Scholar
42. Baselga, J, et al. Phase I studies of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination with cisplatin. J Clin Oncol 2000; 18 (4): 904–914.Google Scholar
43. Cohenuram, M, Saif, MW. Panitumumab the first fully human monoclonal antibody: from the bench to the clinic. Anticancer Drugs 2007; 18 (1): 7–15.Google Scholar
44. Rowinsky, EK, et al. Safety, pharmacokinetics, and activity of ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J Clin Oncol 2004; 22 (15): 3003–3015.Google Scholar
45. Lobo, ED, Hansen, RJ, Balthasar, JP. Antibody pharmacokinetics and pharmacodynamics. J Pharm Sci 2004; 93 (11): 2645–2668.Google Scholar
46. D'Angelica, MI, et al. Phase II trial of hepatic artery infusional and systemic chemotherapy for patients with unresectable hepatic metastases from colorectal cancer: conversion to resection and long-term outcomes. Ann Surg 2015; 261 (2): 353–360.Google Scholar
47. Kemeny, NE, et al. Randomized phase II trial of adjuvant hepatic arterial infusion and systemic chemotherapy with or without bevacizumab in patients with resected hepatic metastases from colorectal cancer. J Clin Oncol 2011; 29 (7): 884–889.Google Scholar
48. Kemeny, NE, et al. KRAS mutation influences recurrence patterns in patients undergoing hepatic resection of colorectal metastases. Cancer 2014; 120 (24): 3965–3971.Google Scholar
49. Brody, T. Introductions and definitions. In Wecker, L, Minneman, KP, eds. Brody's Human Pharmacology, Philadelphia: Elsevier Mosby, 2005; pp. 3–8.
50. Somogyi, A. Clinical pharmacokinetics and issues in therapeutics. In Wecker, L, Minneman, KP, eds. Brody's Human Pharmacology. Philadelphia: Elsevier Mosby, 2005; pp. 41–56.
51. Hollenberg, P. Absortion, distribution, metabolism, and elimination. In Wecker, L, Minneman, KP, eds. Brody's Human Pharmacology.Philadelphia: Elsevier Mosby, 2005; pp. 27–39.
52. Minneman, KP. Receptors and concentration–response relationships. In Wecker, L, Minneman, KP, eds. Brody's Human Pharmacology. Philadelphia: Elsevier Mosby, 2005; pp. 9–25.

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
×