nonsurgical management of recurrent colorectal cancer

15
4278 Nonsurgical Management of Recurrent Colorectal Cancer David J. Vaughn, M.D., and Daniel G. Huller, M.D. Most patients with colorectal carcinoma undergo at- tempts at curative surgery. However, some present with metastatic disease and many others ultimately relapse. Most recurrences of colorectal cancer are not resectable and require nonsurigical approaches such as chemother- apy and radiation therapy directed against local recur- rences, hepatic metastases, and widely disseminated dis- ease. Nonsurgical therapy for locoregional recurrence of rectal cancer can offer significant palliation. Intraarte- rial chemotherapy for liver metastases increases the like- lihood of response compared to systemic treatments, but has little effect on survival. Extrahepatic progression and hepatic toxicity are important limitations to this re- gional therapy. 5-fluorouracil (5-FU) is the mainstay of systemic chemotherapy, and efforts to modulate biochem- ically the cytotoxic effects of 5-FU with folinic acid, phosphonacetyl-L-aspartate, interferon, and other agents have resulted in promising response rates. The different approaches to biochemical modulation are being studied in ongoing cooperative group trials. Novel approaches, including monoclonal antibody therapy, biologic modi- fier therapy, and gene therapy, are under investigation. Cancer 1993; 71:4278-92. Key words: colorectal cancer, metastatic disease, chemo- therapy, radiation therapy. Each year, more than 150,000 Americans receive a diagnosis of colorectal cancer.' Although 80% of pa- tients initially have complete resection of the primary tumor, nearly half have invasion of the cancer through the serosa or regional lymph node involvement, predis- Presented at the American Cancer Society Workshop on the Pretreatment Diagnostic Management of Colorectal Cancer and Fol- low-up Management of Recurrent Disease, San Francisco, California, October 30-31, 1992. From the Hematology/Oncology Division, Department of Medi- cine of the University of Pennsylvania School of Medicine and the University of Pennsylvania Cancer Center, Philadelphia, Pennsyl- vania. Address for reprints: Daniel G. Haller, M.D., University of Pennsylvania Cancer Center, 6 Penn Tower, 3400 Spruce Street, Phil- adelphia, PA, 19104. Accepted for publication February 24, 1993. posing them to a high risk of recurrence.' Recurrent cancer of the large intestine continues to make this dis- ease an important cause of cancer-related death, second only to lung cancer.' Patients with recurrent colorectal cancer can be divided into three groups: those with lo- cal or regional recurrences; those with hepatic metas- tases; and those with widely disseminated disease. His- torically, locoregional failure has been an especially im- portant problem in rectal cancer because of its frequency and because of its association with signifi- cant morbidity. Extrapelvic colon cancer also may be associated with locoregional recurrence, but is more likely to present with distant failure, with the liver be- ing the most common initial single site of metastasis. Although isolated resectable recurrences do occur, non- surgical therapies often are the only options in the man- agement of each category of recurrence of colorectal cancer. Recurrence of colorectal cancer after complete sur- gical resection is thought to be caused by progression of residual occult microscopic disease present at the time of initial surgery. The recently published results of trials of effective postoperative adjuvant therapy have shown decreased recurrence and improved survival for colon and rectal cancer. The recently updated Inter- group tial of adjuvant 5-fluorouracil(5-FU) plus leva- misole in colon cancer demonstrated a 39% decrease in the risk of recurrence, 32% decrease in the risk of cancer-related deaths, and a 31% decrease in the risk of overall deaths in patients with regional lymph node in- volvement (Stage C) at a median follow-up of 5 years.3r4 In two trials of adjuvant therapy of high-risk (Stage B,-C) rectal cancer, the combination of postoperative radiation therapy with 5-FU-based systemic chemo- therapy has been demonstrated to significantly de- crease recurrence rate and the time to recurrence and to improve overall survival compared with a treatment of surgery alone or postoperative radiation therapy An interesting emerging issue is how adjuvant therapy, the standard of care for Stage C colon and high-risk rectal cancer, will affect the natural history of these diseases with respect to changing patterns of re-

Upload: david-j-vaughn

Post on 06-Jun-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

4278

Nonsurgical Management of Recurrent Colorectal Cancer David J. Vaughn, M.D. , and Daniel G. Huller, M.D.

Most patients with colorectal carcinoma undergo at- tempts at curative surgery. However, some present with metastatic disease and many others ultimately relapse. Most recurrences of colorectal cancer are not resectable and require nonsurigical approaches such as chemother- apy and radiation therapy directed against local recur- rences, hepatic metastases, and widely disseminated dis- ease. Nonsurgical therapy for locoregional recurrence of rectal cancer can offer significant palliation. Intraarte- rial chemotherapy for liver metastases increases the like- lihood of response compared to systemic treatments, but has little effect on survival. Extrahepatic progression and hepatic toxicity are important limitations to this re- gional therapy. 5-fluorouracil (5-FU) is the mainstay of systemic chemotherapy, and efforts to modulate biochem- ically the cytotoxic effects of 5-FU with folinic acid, phosphonacetyl-L-aspartate, interferon, and other agents have resulted in promising response rates. The different approaches to biochemical modulation are being studied in ongoing cooperative group trials. Novel approaches, including monoclonal antibody therapy, biologic modi- fier therapy, and gene therapy, are under investigation. Cancer 1993; 71:4278-92.

Key words: colorectal cancer, metastatic disease, chemo- therapy, radiation therapy.

Each year, more than 150,000 Americans receive a diagnosis of colorectal cancer.' Although 80% of pa- tients initially have complete resection of the primary tumor, nearly half have invasion of the cancer through the serosa or regional lymph node involvement, predis-

Presented at the American Cancer Society Workshop on the Pretreatment Diagnostic Management of Colorectal Cancer and Fol- low-up Management of Recurrent Disease, San Francisco, California, October 30-31, 1992.

From the Hematology/Oncology Division, Department of Medi- cine of the University of Pennsylvania School of Medicine and the University of Pennsylvania Cancer Center, Philadelphia, Pennsyl- vania.

Address for reprints: Daniel G. Haller, M.D., University of Pennsylvania Cancer Center, 6 Penn Tower, 3400 Spruce Street, Phil- adelphia, PA, 19104.

Accepted for publication February 24, 1993.

posing them to a high risk of recurrence.' Recurrent cancer of the large intestine continues to make this dis- ease an important cause of cancer-related death, second only to lung cancer.' Patients with recurrent colorectal cancer can be divided into three groups: those with lo- cal or regional recurrences; those with hepatic metas- tases; and those with widely disseminated disease. His- torically, locoregional failure has been an especially im- portant problem in rectal cancer because of its frequency and because of its association with signifi- cant morbidity. Extrapelvic colon cancer also may be associated with locoregional recurrence, but is more likely to present with distant failure, with the liver be- ing the most common initial single site of metastasis. Although isolated resectable recurrences do occur, non- surgical therapies often are the only options in the man- agement of each category of recurrence of colorectal cancer.

Recurrence of colorectal cancer after complete sur- gical resection is thought to be caused by progression of residual occult microscopic disease present at the time of initial surgery. The recently published results of trials of effective postoperative adjuvant therapy have shown decreased recurrence and improved survival for colon and rectal cancer. The recently updated Inter- group tial of adjuvant 5-fluorouracil(5-FU) plus leva- misole in colon cancer demonstrated a 39% decrease in the risk of recurrence, 32% decrease in the risk of cancer-related deaths, and a 31% decrease in the risk of overall deaths in patients with regional lymph node in- volvement (Stage C) at a median follow-up of 5 years.3r4

In two trials of adjuvant therapy of high-risk (Stage B,-C) rectal cancer, the combination of postoperative radiation therapy with 5-FU-based systemic chemo- therapy has been demonstrated to significantly de- crease recurrence rate and the time to recurrence and to improve overall survival compared with a treatment of surgery alone or postoperative radiation therapy

An interesting emerging issue is how adjuvant therapy, the standard of care for Stage C colon and high-risk rectal cancer, will affect the natural history of these diseases with respect to changing patterns of re-

Nonsurgical ManagementlVaughn and Haller 4279

currence. For example, 5-FU/levamisole adjuvant treatment in Dukes C colon cancer resulted in a de- creased recurrence rate at all disease sites but with rela- tively greater improvement in extraabdominal region^.^ An additional issue is whether aggressive postoperative monitoring, including surveillance by carcinoem- bryonic antigen (CEA) levels and imaging techniques such as computed tomography (CT), magnetic reso- nance imaging, and radioimmunoscintography will re- define the natural history of recurrent colorectal cancer and allow earlier detection and treatment of recurrent disease.

Locoregional Recurrence of Rectal Cancer

Locoregional recurrence of rectal cancer is an important cause of morbidity and mortality. The risk of pelvic re- currence is directly related to the stage of the tumor. The incidence of local recurrence of rectal cancer with extension through the wall (Modified Astler-Coller (MAC), B2 and B3; TNM, T,N, and T,N,) or with re- gional lymph node involvement (MAC, C1; TNM, T,N, or T,N,) has been reported to be 20-40%.8,9 Lesions with both of these poor prognosis characteristics (MAC, C2 or C3; TNM, T,-,, N1.J are associated with an over- all recurrence rate of 40-65%.8*9 In addition, there is a relationship between the risk of local failure and the location of the tumor within the rectum; that is, lower segment cancers are much more likely to recur than middle or upper segment lesions." Local failure in pa- tients with rectal cancer may be related to the inability to obtain wide radial surgical margins around the pri- mary tumor when there is infiltration of the tumor through the intestinal wall into perirectal fat. In addi- tion, it may not be possible to adequately excise all of the lymph node regions at risk for recurrence, especially with lower segment tumors. Factors that have not been shown to clearly affect the rate of recurrence include the type of surgery (that is, abdominoperitoneal versus sphincter-sparing resection) and the extent of clear lon- gitudinal surgical margins."

When a pelvic recurrence of rectal cancer occurs, an

Table 1. Management of Locoregional Recurrence of Rectal Cancer

aggressive multimodality approach is warranted be- cause of potentially devastating symptoms, such as pain and organ obstruction." If a pelvic recurrence is amenable to surgical resection, this may represent the best option. An aggressive combined modality ap- proach of surgically resectable, locally recurrent rectal cancer has been reported from the Mayo Clinic; the approach incorporates surgical resection andintraopera- tive radiation therapy, followed by postoperative exter- nal beam treatment. In this highly selected group of 24 patients, a local control rate of 85% and prolonged sur- vival of 67% were seen at a mean follow-up of 20 months.', However, most series have shown that most locoregional pelvic recurrences are unresectable, partic- ularly when sympt~matic. '~ Thus, management of unresectable locoregional recurrence with nonsurgical techniques represents the most practical and widely used palliative treatment option (Table 1).

Radiation therapy historically has been the most important modality in the management of symptomatic pelvic recurrences. In retrospective series, this therapy has been associated with subjective improvement, measured by initial pain control and improved quality of life, in 80-90% of patients.15,16 However, long-term prolongation of disease-free and overall survival after palliative radiation alone is u n ~ o m m o n . ~ ~ * ~ ~ Prospective series have examined the role of treatment with exter- nal beam radiation therapy alone and in combination with 5-FU-based chemotherapy in patients with locally recurrent, primary locally advanced, and residual rectal

For example, a Radiation Therapy Oncol- ogy Group (RTOG) trial randomized patients to receive external beam radiation alone (4500-5100 rads/5-6 weeks with or without boost) or radiation therapy with concurrent 5-FU, followed by maintenance 5-FU and semustine (methyl-CCNU)." Sixty-four percent of the entire group ultimately experienced local failure, with no significant difference in disease control seen be- tween the two arms. Median survival between the two treatment arms was not significantly different (in the radiation therapy alone group: 17 months; in the combined therapy group: 18 months). A nonsta-

No. of Reference Type patients Treatment Response rates Survival (mo)

DobrowskyI5 R 58 EBR 90% improved pain Median 19.8 2 yr: 29% Pacini16 R 143 EBR 80.4% improved pain 2 yr: 17.5% R~rninger'~ P 129 EBR versus EBR/CT NR Median 17 versus 18

Danjoux'* P 30 EBR/CT 10% objective regression Median 17 Dobrowsky" P 15 EBR/CT 47% objective regression Median 14 R: retrospective; P: prospective; EBR external beam radiation; C T chemotherapy; NR not reported.

2 yr: 36% versus 44%

4280 CANCER Supplement Iune 15, 1993, Volume 71, No. 12

tistically significant trend toward improved survival at 2 years was seen in the combined group (44% ver- sus 36%).

A second prospective trial, by the Eastern Coopera- tive Oncology Group (ECOG), also examined the role of chemoradiation therapy for residual, recurrent, or inoperable rectal cancer.” Patients received either con- tinuous radiation plus 5-FU or split-course radiation plus 5-FU, and some also received postradiation mainte- nance 5-FU plus methyl-CCNU. An overall objective response rate was observed in only 10% of patients with measurable disease; median survival in each group was 17 months. In both of these prospective trials, toxicity was substantial, especially in patients treated with combined modality therapy. Intestinal complications, including severe diarrhea, small intes- tinal obstruction, and perforation, were the most im- portant causes of treatment-related morbidity. In fact, the ECOG trial was terminated because of excessive late treatment toxicity, primarily intestinal complications.’* In a recently reported series of 15 patients with inopera- ble presacral recurrences of rectal cancer, split-course radiation therapy was administered with 5-FU/mito- mycin C. This nonrandomized prospective trial showed an objective response rate of 47%, with a median sur- vival of 14 months. Only 6 of 15 patients completed therapy as planned because of excessive gastrointesti- nal and hematologic toxicity.”

Because of the heterogeneous patient populations treated in these prospective studies, definitive recom- mendations with respect to the optimum management of pelvic recurrences can not be concluded. Given the proven efhcacy of combined chemoradiation therapy in the adjuvant setting of rectal ~ a n c e r , ~ - ~ this has become the standard treatment of local recurrence in patients who have not previously been radiated. However, as demonstrated in the prospective trials of combined mo- dality therapy, toxicity may be substantial. As increas- ing numbers of patients receive aggressive adjuvant chemoradiation therapy after the primary surgical treatment of rectal cancer, fewer patients will have local failure. When patients who received radiation therapy experience disease recurrence, additional radiation ther- apy may be precluded and thus treatment may be lim- ited to chemotherapy alone with other palliative mea- sures.

Pelvic arterial chemotherapy given with 5-FU plus mitomycin C administered via the bilateral iliac arteries has been studied in patients with painful pelvic recur- rences of rectal cancer who have not responded to prior radiation therapy or systemic chemotherapy. In one se- ries, 50% of these patients experienced short-term (3-4 months) pain relief, and therapy was associated with a median survival of 12 months.” Thus, this form of ther-

apy may not be cost-effective given its technical limita- tions and short duration of benefit and is not routinely recommended.22 Intraluminal cryotherapy is an innova- tive therapeutic modality that may be useful for pallia- tive treatment of recurrent rectal cancer. In a pilot se- ries, 20 patients with unresectable local recurrence re- ceived cryoprobe treatment of the intraluminal tumor. Nine patients experienced complete symptom relief, with a median symptom-free interval of 11 months; 5 of these 9 patients died without evidence of local dis- ease.23

In addition to specific antitumor therapy, symptom- atic pelvic recurrences often may require additional pal- liative therapy for control of pain. Lumbar-sacral nerve block and the use of opioid analgesics, especially of the long-acting type, may be important treatment modali- ties used in conjunction with antitumor therapies when the patient has experienced disease progression after radiation and chemotherapy.22 In addition, noncurative surgical procedures such as the placement of ureteral stents and colostomies in patients with organ obstruc- tion caused by pelvic recurrence may offer significant palliation in selected patients.

Liver Metastases

The liver is the most common site of initial distant re- lapse in colorectal cancer. Although approximately 20% of liver metastases are identified synchronously with presentation of the primary tumor, most become clinically apparent after surgical r e ~ e c t i o n . ~ ~ , ~ ~ Survival of patients with liver metastases caused by colorectal cancer is extraordinarily variable. The commonly ac- cepted median survival of 6 months may be shorter or much longer, depending on major prognostic variables for predicting survival, such as the extent of hepatic involvement, presence or absence of extrahepatic dis- ease, level of abnormalities of liver function tests, and performance status of the patient.26 Although retrospec- tive and prospective series have shown surgical resec- tion to be the only potentially curative approach for the treatment of liver metastases, most patients do not have disease that is resectable. Only approximately 20% of patients who undergo resection are alive and free of disease at 5 year^.^^,^' Thus, nonsurgical modalities re- main important in the management of patients with colorectal liver metastases. Standard systemic chemo- therapy with 5-FU or 5-FU/leucovorin may be ex- pected to result in objective response rates of 25-4070 in these patients.29 To potentially increase this observed response rate, organ-targeted arterial chemotherapy has been developed to improve the efficacy of treat- ment for patients with tumor localized to the liver.

The rationale for intraarterial chemotherapy is

Nonsurgical ManagementlVuughn and Huller 4281

based on the hypothesis that increased local drug con- centration may improve the tumor response rate.30 Whereas normal liver parenchyma and early microme- tastases are fed by the portal vein, established hepatic metastases are believed to receive most of their blood supply from the hepatic artery.31 Thus, intraarterial he- patic chemotherapy results in higher tumor drug con- centration than that achieved with intravenous adminis- t ra t i~n.~ ' In addition, metabolism of agents, including the fluoropyrimidines, delivered via the hepatic intraar- terial route results in the clearance of at least 80% of the administered dose. This allows less exposure of normal extrahepatic tissues to the drug, resulting in less sys- temic Thus, there is compelling physiologic and pharmacologic rationale for the administration of drugs via the hepatic artery."

In the 1970s, investigators began trials using exter- nal pumps to deliver 5-FU and floxuridine via the cath- eterized hepatic artery to patients with liver metastases from a variety of gastrointestinal adenocarcinomas, in- cluding colorectal ~ a n c e r . ~ ~ - ~ ~ These early studies dem- onstrated objective response rates ranging from 25% to 71%, with an average response rate of 55%, suggesting an improvement over standard 5-FU chemotherapy. However, this treatment often required hospital admit- tance and immobilization of the patient. Arterial throm- bosis (go/, on average) and catheter dislodgement (in 10-15% of patients) also were important complica- t i o n ~ . ~ ~ For these reasons, hepatic arterial therapy was not widely accepted by the oncologic community, de- spite the increased response rates.

The field of arterial-directed chemotherapy for liver metastases was advanced by the development of surgically implanted infusion pumps.41 These pumps have been shown to have advantages to earlier tech- niques because of the decreased risk of arterial thrombo- sis, catheter dislodgement, and catheter-related sep- s~s .~ ' Cost-benefit analysis comparing these pumps to the external delivery system also has shown the im- plantable pump to be more economical when more than 4 months of treatment is planned.42

Randomized trials (Table 2) of intraarterial fluoro- deoxyuridine via the implantable pump compared with systemic chemotherapy in patients with unresectable hepatic metastases from colorectal cancer have been performed by Kemeny et al.,43 Chang et al.,44 Hohn et al.,45 Martin et al.,46 and Rougier et al.47 Three of the studies used floxuridine as the systemic the rap^:^-^^ whereas two used 5-FU.46,47 All of these studies have shown a statistically significant improved objective re- sponse rate in the intrahepatic group (42-62%) com- pared with the systemic group (10-219'0). However, no definite significant survival advantage for intrahepatic therapy has been demonstrated in any of these studies. In the studies performed by Kemeny et al.43 and Hohn et al.,45 the therapy for patients receiving systemic ther- apy who experienced hepatic progression could be crossed-over to the intrahepatic therapy. Thus, survival advantages of patients for either therapy alone could not reliably be determined.

In the study by Chang et al., a statistically insignifi- cant survival difference of 17 months for the intrahepa- tic group was demonstrated compared with 12 months for the systemic group when analyzed by intent-to- treat. However, if only treated patients were analyzed, the median survival of the intrahepatic group was 19 months compared with 13 months for the systemic

The study by Rougier et al. showed a median survival of 14 months for the patients treated with in- trahepatic therapy, compared with 10 months for those treated with "palliative" therapy (P < 0.02). However, only half of the latter group received systemic 5-FU, whereas the other half received no chemotherapy. The true survival and response rates for the subgroup treated with systemic 5-FU are not de~cribed.~' Thus, while these studies have nearly always demonstrated a clear advantage of intrahepatic therapy for objective response rates, the effect of this treatment on survival is not clear, perhaps because of methods of study design and data analysis. No randomized trial has compared intrahepatic floxuridine with systemic 5-FU/leucovorin regimens that have been associated with increased re-

Table 2. Randomized Trials of Intrahepatic Floxuridine versus Systemic Floxuridine or 5-Fluorouracil

No. of Response Median survival patients rate (mo)

Reference IH s IH S P value IH S P value

kern en^'^ 48 50 50% 20% 0.001 17 12 0.4 Chang" 21 29 62% 17% < 0.003 17 12 0.27 Hohn" 50 65 42% 10% = 0.0001 16.7 16.1 NS Martin'6 33 36 54% 21% = 0.02 12.6 10.5 0.53 Rougier4' 81 82* 48% 14% NS 14 10 < 0.02 IH: intrahepatic; S: systemic; NS: not stated. * Onlv half of these patients received 5-fluorouracil

4282 CANCER Supplement June 25, 2993, Volume 71, No. 12

sponse rates and survival advantage compared with the use of 5-FU alone.29

There are two major drawbacks to intrahepatic ther- apy for the treatment of liver metastases from colorectal cancer: extrahepatic progression and toxicity. Despite clearly improved intrahepatic tumor response rates and a trend toward improved survival in some trials with intrahepatic chemotherapy, a review of these data re- veals that consistent improvements in overall and dis- ease-free survival are limited by the development of distant metastasis in extrahepatic sites. Extrahepatic re- lapse has been reported to occur in 36-50% of patients treated with intrahepatic therapy, most commonly in the lungs.22 Because symptoms and survival are most likely to be determined by the status of hepatic disease, continued intrahepatic therapy occasionally has been recommended, even when extrahepatic progression is present.22 Others have attempted to delay or prevent extrahepatic failure by initiating trials of combined in- trahepatic and systemic chemotherapy.

Initial studies from Memorial Sloan-Kettering Cancer Center using combinations of intrahepatic flox- uridine and systemic 5-FU/leucovorin have suggested increased gastrointestinal and hepatic toxicity when the systemic chemotherapy regimen was given for 5 succes- sive days and when the systemic chemotherapy regi- men closely followed the intrahepatic treatment4* A partial response rate of 56% with a median survival of 16 months was noted in 21 patients treated in this trial. A trial by Safi49 has been reported in which patients with liver metastases were randomized to receive either floxuridine 0.2 mg/kg/day by the intrahepatic route or floxuridine 0.3 mg/kg/day administered simulta- neously by the intrahepatic and systemic routes for 14 days each month. A response rate of 59% was reported in the intrahepatic group and 57% in the combined group. Seventy-nine percent of the intrahepatic and 51 9'0 of the combined group experienced extrahepatic relapse in a median follow-up of 24 months ( P < 0.01). No significant survival differences were observed, and toxicity in both treatment arms was similar.49

At the University of Pennsylvania, a Phase 1/11 trial of combined intrahepatic floxuridine with weekly sys- temic 5-FU plus high-dose leucovorin is being piloted for patients with liver metastases caused by colorectal cancer. In this trial, the weekly systemic and intrahepa- tic regimens are separated by rest periods in an attempt to prevent the gastrointestinal, mucosal, and hepatic toxicity reported in the Memorial Sloan-Kettering Cancer Center pilot study. Whether combined intrahe- patic and systemic treatment will prove to offer advan- tages to the use of either treatment modality alone re- mains to be determined.

The second major limitation to intraarterial floxuri-

dine is hepatobiliary toxicity. In a recent review, approx- imately 25% of patients who received intrahepatic flox- uridine had bilirubin elevation, with biliary sclerosis oc- curring in 159'0.~' Biliary sclerosis is observed most often when high doses of floxuridine (0.3 mg/kg or greater) are infused for 2 or more weeks or when early signs of biliary toxicity, such as an increasing alkaline phospha- tase level, are ignored. Chemical hepatitis also may oc- cur and should be suspected when transaminases are increased. Both of these hepatobiliary toxicities often resolve with temporary cessation of treatment. Perma- nent damage to the biliary tree has been reported."

Innovative approaches to limit hepatobiliary toxic- ity have been studied. One such approach has been to alternate intrahepatic floxuridine with 5-FU, an agent with less first-pass hepatic extraction and thus less bili- ary toxicity than floxuridine, but also less effi~acy.~' Al- ternating intrahepatic floxuridine 0. l mg/kg/day for 7 days with intrahepatic bolus 5-FU 15 mg/kg on days 14, 21, and 28 resulted in response rates of 51%, with only approximately 10% of patients requiring dose modification because of hepatobiliary toxicity; this rate is less than that previously demonstrated in control subjects. An alternate approach was proposed by Seifer et al., who conducted a prospective trial in which intra- hepatic floxuridine 0.3 mg/kg/day for 2 weeks per month was administered with or without intrahepatic dexamethasone at a total dose of 20 mg during a 2- week period. A trend toward less biliary toxicity, as manifested by decreased bilirubin levels, was noted in the group receiving the dexamethasone, but the differ- ence was not significant (99'0 versus 32%; P = 0.07).52

Circadian modification (CM) of intrahepatic flox- uridine infusion has been compared with flat drug ad- ministration in a retrospective study of 50 patients at the University of Minnesota.53 By administering the drug via CM with 68% of the drug administered in the early evening (between 3 p.m. and 9 p.m.) and only 3% in the morning (between 3 a.m. and 9 a.m.), the group receiving the CM floxuridine tolerated more of the drug with less hepatic toxicity than did control subjects re- ceiving standard floxuridine. Although innovative ap- proaches to administering floxuridine by the intrahepa- tic route may reduce hepatic toxicity, none have been demonstrated to be uniformly reliable in a prospective randomized trial, and use of these regimens outside the context of a clinical trial cannot be recommended.

An additional toxicity related to intrahepatic ther- apy is gastric ulceration, which is reported in 40% of patient^.^' This presumably is attributable to perfusion of the stomach and the duodenum with floxuridine via the small vessels branching from the hepatic artery. The incidence of this toxicity may be markedly decreased by careful surgical techniques, including ligation of the

Nonsurgical Management/Vaughn and Haller 4283

vessels that supply the stomach and duodenum.54 Gas- tric ulceration often resolves upon discontinuation of therapy and may respond to H,-antagonist therapy, such as cimetidine or ranitidine.

In an attempt to increase the response rate with intrahepatic therapy, additional nonrandomized stud- ies have used floxuridine in combination with other agents administered intraarterially, including mitomy- cin C or platin~m,’~-’~ 5-FU,” or l e u c ~ v o r i n . ~ ~ ~ ’ ~ These studies generally have not shown a dramatic difference in the rate of response compared with that seen with floxuridine alone. In addition, hepatobiliary toxicity was markedly increased in the floxuridine/leucovorin trial^.'^,^^ Additional studies combining floxuridine with other modulators (alpha-interferon) or using intra- arterial floxuridine in the adjuvant mode after curative resection of liver metastases are planned.” Finally, in- novative Phase I trials have used the intraarterial route for delivering biologic agents to the liver, including in- terleukin-2 (IL-2)60 and tumor necrosis factor (TNF).61 Although IL-2 produced an overall response rate of less than lo%, TNF showed some promise in patients who had failed to respond to standard chemotherapy, producing an objective response rate of approxi- mately 20%.

Hepatic arterial chemoembolization has been used in the management of liver tumors. This therapy in- volves instillation into the hepatic artery of an embolic mixture that incorporates chemotherapeutic agents.’* By blocking perfusion of the tumor, and by delivering high concentrations of cytotoxic agents directly to the tumor, chemoembolization has resulted in significant response rates in metastatic carcinoid tumors6’ and pri- mary hepatocellular carcinoma^.^^ Recently, chemoem- bolization trials have been expanded to include hepatic metastases caused by colorectal ~ a n c e r . ~ ~ - ~ ~ The largest series of patients treated with this technique included 52 patients with hepatic metastases from colorectal car- cinoma who had failed to respond to systemic chemo- therapy.66 In this trial, patients were treated with an embolic mixture of collagen, cisplatin, doxorubicin, and mitomycin C. Thirty-four percent of patients had a more than 50% reduction of measurable tumors on CT, and 78% had a more than 50% reduction in CEA levels. Projected median survival after embolization was 335 days.

Another experimental approach to the directed treatment of liver metastases is percutaneous ethanol injection into the tumor.67 In one series, 14 patients, half with metastatic colorectal cancer, were treated in 175 sessions for a total of 204 ethanol injections with- out serious complications. In the patients with colorec- tal metastases, a decline in CEA was noted in six of seven patients. However, significant responses based

on CT criteria were not seen. The best responses in this series were seen in patients with metastases from neuro- endocrine tumors. ”

Investigators at the University of Pittsburgh have reported therapy of unresectable liver metastases from a variety of cancers with the intraoperative placement under ultrasonographic guidance of cryoprobes that subject the tumor to temperatures as low as -180°C. Preliminary results demonstrate that 36 of 53 (68%) patients treated with ”cryosurgery” are alive 1-39 months after treatment (average, 12 months), with nor- malization of CT and CEA levels occurring in 29 pa- tients. 68

Although these innovative liver-directed therapies offer promise of improved local control, the reported nonrandomized series may reflect positive outcomes in highly selected patient cohorts and may not be general- izable. In addition, the progression of untreated hepatic micrometastases and extrahepatic metastatic progres- sion during treatment with these local therapies may limit their efficacy in terms of overall survival.

In summary, randomized studies have shown that the intraarterial route for administering chemotherapeu- tic agents to liver metastases results in a greater re- sponse rate than that seen with systemic chemother- apy. However, intraarterial therapy has not been di- rectly compared with systemic 5-FU/leucovorin in a randomized trial. In addition, survival has not defini- tively been shown to be significantly improved by this therapy. Dose-limiting toxicity, principally hepatobili- ary, also affects the widespread use of this treatment. Trials that combine intrahepatic therapy with systemic chemotherapy define ways in which extrahepatic fail- ure may be reduced. Finally, chemoembolization, percu- taneous ethanol injection, and cryosurgery are innova- tive options that need additional testing in clinical trials as palliative therapy for patients with nonresectable symptomatic liver metastases who have failed primary systemic chemotherapy. As local measures, these tech- niques do not fully address the natural history of recur- rent colorectal cancer, which usually is multicentric. Combinations of regional and systemic treatments may allow more comprehensive therapy for these patients.

Disseminated Disease

Since the mid-l950s, the mainstay of systemic chemo- therapy for metastatic colorectal cancer has been 5-FU. In collected series of patients, objective response rates obtained with standard bolus 5-FU have been in the range of 10-20%, with no consistent improvement in survival.69 Mitomycin C and the chloroethylnitroso- ureas have shown only marginal activity in patients

4284 CANCER Supplement June 15, 2993, Volume 71, No. 12

with advanced colorectal cancer, whereas other chemo- therapeutic agents, alone or in combination with 5-FU, generally have been ineff e ~ t i v e . ~ ~ The most important recent advance in the management of metastatic colo- rectal cancer has been the evolution of programs that use biochemical modulation of 5-FU.29 Biochemical modulation refers to the alteration of 5-FU cytotoxicity by the addition of other agents, such as leucovorin (fo- linic acid), recombinant interferon alpha-2a (rIFN a-2a), phosphonacetyl-L-aspartate (PALA), methotrex- ate, and cisplatin.The tumor cytotoxicity of 5-FU depends on alteration in a number of biochemical pathways involved in nucleotide synthesis. In one im- portant pathway, the metabolite of 5-FU, fluorodeoxy- uridine monophosphate (FdUMP), competitively binds to thymidylate synthase, resulting in diminished DNA synthesis by the depletion of thymidine. 5-FU also is metabolized via the ribonucleotide pathway to yield 5- fluorouridine triphosphate (FUTP), a competitive sub- strate for RNA polymerase that upon incorporation into RNA may be cytot~xic.~' The various modulators of 5- FU act at different points of the metabolic cycle of 5-FU to enhance its cytotoxicity (see Fig. 1).

The best-studied modulator of 5-FU is leucovorin, a reduced folate that enhances the cytotoxicity of 5-FU by increasing the inhibition of thymidylate synthase by FdUMPe7' Multiple clinical trials recently have been re- ported that establish the efficacy of various combina- tions of 5-FU/leucovorin in metastatic colorectal cancer compared with standard bolus 5-FU therapy (Table 3).29 Petrelli et aL7' from Roswell Park Cancer Institute (RPCI) reported a treatment program in which 5-FU 600 mg/m2 plus leucovorin 500 mg/m2 (high dose) were administered weekly for 6 weeks, followed by a

2-week rest period. In a Phase I11 trial comparing this regimen to single-agent 5-FU, a statistically significant improvement in objective response rates was seen (48% versus 11%; P = 0.0009).

The efficacy of this regimen was additionally con- firmed in a Phase I11 trial by the Gastrointestinal Tumor Study Group (GITSG), in which the same weekly 5-FU plus high-dose leucovorin regimen was compared with weekly 5-FU plus low-dose (25 mg/m2) leucovorin and with 5-FU alone.72 A statistically significant improved response rate of 30% was seen with the high-dose leu- covorin treatment arm (P < 0.01) but not with the low- dose leucovorin regimen. Survival was modestly, but not statistically significantly, prolonged. Toxicity with the high-dose leucovorin regimen was substantial. Se- vere diarrhea occurred in 25% of those treated, and 5% of patients died of diarrhea-related events. The investi- gators advised careful monitoring of patients treated with this regimen, early treatment of diarrhea-related dehydration with intravenous fluids, and cessation of chemotherapy until resolution of all symptoms. Al- though this weekly 5-FU/high-dose leucovorin regi- men also occasionally produced mucositis and conjunc- tivitis, significant hematologic toxicity was uncommon. Because of the severe diarrhea associated with this regi- men, standard practice reduces the dose of 5-FU to 500 mg/m2 given with leucovorin 500 mg/m2.

A second commonly used regimen of 5-FU/leuco- vorin was studied prospectively by the North Central Cancer Treatment Group (NCCTG).73 This program ini- tially incorporated 5-FU 370 mg/m2 with leucovorin 200 mg/m2 (high dose) or 20 mg/m2 (low dose) admin- istered for 5 consecutive days monthly. Because of lack of significant toxicity in

IFN LV > FH2 FH4

Thyrnidylate Synthase

5-FdUMP -5-FdUDP -5-FdUTP *5-FDNA

IFN P MTX P 5-FUdR

5-FU -< MTxx 5-FUMP -> J-FUDP,-D 5-FUTP -> 5-FRNA

PALA

the- first group of 100 patients,

Figure 1. Pathways of 5-FU metabolism and the effects of biochemical modulators. After initial phosphorylation of 5-FU to 5-FUMP, additional metabolism along the ribonucleotide pathway yields 5- FUTP, a substrate for RNA polymerase (RNAse); incorporation of 5-FU into RNA may be cytotoxic. Phosphonacetyl-L-aspartate (PALA) inhibits de novo pyrimidine synthesis, depleting UTP, and increasing 5-FU incorporation into RNA. 5-FU also may be metabolized along the deoxynucleotide pathway. 5-FdUMP inhibits thymidylate

~~

synthase (TS) and arrests DNA synthesis. Leucovorin (LV) increases intracellular levels of reduced folates and increases inhibition of thymidylate synthase (TS). Interferon-a (IFN) inhibits thymidylate synthase (TS) and augments conversion of 5-FU to 5-FdUMP. Methotrexate (MTX) decreases purine nucleotide synthesis, resulting in an increase of intracellular phosphoribosylpyrophosphate, favoring the synthesis of 5-FdUMP and FUTP.

Nonsurgical Management/Vaughn and Huller 4285

Table 3. Selected Phase 111 Trials of 5-Fluorouracil(5-FU)/Leucovorin

No. of Reference uatients Dosehchedule Response rate

Median survival

Petrelli7' 44 5-FU 600 mg/m2 + L 500 mg/m2 X 6 wk versus 5-FU 450 mg/m2 X 5 every 4 wk

GITSG72 318 5-FU 600 mg/m2 + L 500 or 25 mg/m2 X 6 wk versus 500 mg/m2 X 5 every 4 wk

mg/m2 X 5 d every 4 wk versus 5FU 500 mg/mz X 5 d every 4 wk

5-FU 370 mg/m2 X 5d + L 500 mg/m2 X 6 d every 4 wk versus 5-FU 370 mg/m2 X 5 d every 4 wk

5-FU 370 mg/m2 + L 200 mg/m2 X 5 d every 4 wk versus 5-FU 370 mg/m2 X 5 d every 4 wk

5-FU 400 mg/m2 + L 200 mg/m2 X 5 d every 4 wk versus 5-FU 480 mg/m2 X 5 d then 600 mcr/m2/wk

NCCTG73 208 5-FU 370-425 mg/m2 + L 20 or 200

D o r o ~ h o w ~ ~ 74

E h r l i ~ h m a n ~ ~ 124

valone7' 153

48% versus 1 1 % (P = 0.0009)

30.3% versus 18.8% versus 12.1% (P < 0.01 for high- dose versus 5-FU alone)

43% versus 26% versus 10% (P = 0.001 for low-dose versus 5-FU alone)

44% versus 13% (P = 0.0019)

33% versus 7% ( P < 0.0005)

18.8% versus 16.9% (P < 0.4)

Estimated 12 mo versus 11 mo (P = 0.6)

55, 45,46 wk (P = 0.08)

53,52, 34 wk (P = 0.05)

62 versus 55 wk (P = 0.25)

54 versus 41 wk (P = 0.05)

24 versus 20 wk (P > 0.4)

L leucovorin; NS: not significant

the investigators increased the 5-FU dose to 425 mg/mz in the low-dose leucovorin treatment arm. These two regimens were compared prospectively to 5-FU alone and to 5-FU in combination with methotrexate and with cisplatin. The low-dose leucovorin schedule pro- duced the highest response rate (43%) compared with the high-dose regimen (26%) or with 5-FU alone (1Oo/o),

In addltion, treatment with either of the leucovorin regi- mens resulted in a modest, but statistically significant, prolongation of survival compared with 5-FU alone (12.2 months for the high-dose regimen, 12.0 months for the low-dose regimen, and 7.7 months for the 5-FU alone regimen). Quality of life indices, including perfor- mance status, weight gain, and symptoms, were signifi- cantly improved in the low-dose leucovorin treatment arm. Toxicity of the 5-FU/leucovorin regimens was principally stomatitis, occurring severely in 30% of those in the high-dose treatment arm and in 26% of those in the low-dose treatment arm. Severe diarrhea did not occur more often in the leucovorin treatment arms than in the 5-FU alone treatment arms. No deaths were attributed to nonhematologic toxicity. Severe leu- kopenia (leukocyte count of less than 2000) occurred in approximately 20% of patients treated with 5-FU and leucovorin. Thus, because of the increased response rate, acceptable toxicity, and lower cost of the 5-FU/ low-dose leucovorin regimen, the authors and othersz9 have recommended this regimen as the standard treat- ment of metastatic colorectal cancer.

The results of this trial recently were updated, con- firming the efficacy of the 5-day 5-FU with low-dose (20 mg/m2) leucovorin compared with 5-FU and high-

dose leucovorin (200 mg/m2) or a 5-FU/methotrexate ~ombina t ion .~~ As shown in Table 3, other randomized trials of 5-day regimens of 5-FU/leucovorin have been performed, generally using higher doses of leucovorin, ranging from 200 to 500 mg/mz.75-77 In most, statisti- cally significant improved response rates were seen with the combined therapy compared with 5-FU alone. In one trial, a statistically significant increase in survival was reported.76

A direct comparison of the two most commonly used regimens of 5-FU/leucovorin recently was re- ported, in which 370 patients with advanced colorectal cancer were randomized to receive the GITSG regimen (5-FU 600 mg/mz and leucovorin 500 mg/m2 weekly for 6 weeks, followed by a 2-week rest) or the NCCTG regimen (5-FU 425 mg/m2 and leucovorin 20 mg/m2 daily for 5 days every 4-5 weeks).78 No statistically sig- nificant difference was demonstrated between the two regimens with respect to response rate (28% for GITSG, 33% for NCCTG; P = 0.47) or median survival (10 months for both; P = 0.69). Toxicity analysis confirmed diarrhea to be the dose-limiting toxicity of the GITSG regimen, and stomatitis for the NCCTG regimen. Life- threatening and lethal toxicity of the regimens was simi- lar at 2%, but total time in the hospital to manage toxic- ity was greater for the GITSG regimen (436 versus 317 patient-days; P = 0.07). The authors concluded that the NCCTG regimen is superior based on decreased patient hospital stay for toxicity and treatment cost. However, in this trial the dose of 5-FU used in the high-dose leu- covorin regimen was 600 mg/m2, rather than the better tolerated, and more widely used, 500 mg/m2.

4286 CANCER Supplement June 15, 2993, Volume 71, No. 12

A meta-analysis of updated data from ten random- ized clinical trials of 5-FU/leucovorin compared with 5-FU alone has been reported.79 A total of 803 patients were treated with 5-FU/leucovorin regimens and 5 78 with 5-FU alone. Treatment with 5-FU/leucovorin ei- ther on a weekly or 5-day per month regimen produced a highly significant benefit compared with single-agent 5-FU in terms of overall objective response rates (23% versus 11%; P < However, this increased re- sponse rate did not result in improvement in overall survival. The authors attributed the discrepancy be- tween response and survival to the large number of patients in both groups that had no response to therapy and the low complete remission rates of 3% or less in both groups. In addition, data from the NCCTG trial, which demonstrated a survival advantage for patients receiving 5-FU/leucovorin, were not included in the meta-analysis. Many patients with tumor progression after 5-FU therapy alone subsequently received treat- ment with 5-FU/leucovorin, so a true survival compari- son was not possible.

Although progress in treating metastatic colorectal cancer has been made by modulating 5-FU with leuco- vorin, this therapy is not curative and most patients do not achieve an objective response. Thus, efforts have been made in investigating other modulating agents: specifically, rIFN a-2a, PALA, methotrexate, and cis- platin. rIFN a-2a was shown in preclinical investigation to interact with 5-FU by a variety of mechanisms: by decreasing 5-FU clearance, by decreasing thymidine ki- nase activity, by inhibiting thymidine incorporation into DNA, by decreasing the rate of thymidine phos- phorylation,” by enhancing thymidylate synthetase in- hibition,” and by increasing intracellular accumulation of FdUMP.” On the basis of this preclinical data, Wadler et aLa3 treated 30 patients with advanced colo- rectal cancer with 5-FU 750 mg/m2 by continuous infu- sion for 5 consecutive days, followed by weekly bolus administration and rIFN a-2a 9 X lo6 U thrice weekly. Although no major responses were seen in any of the patients who had previously received 5-FU, a response rate of 76% was demonstrated in previously untreated patients.83 Initial enthusiasm for this regimen was tem- pered by a second Phase I1 study that used the same regimen in untreated patients and showed an overall response rate of 35%, with a median duration of re- sponse of 7.5 months. Significant granulocytopenia (43%), mucositis (50%), and a flu-like syndrome of fever, chills, and myalgias seen in nearly all patients were the major to~ici t ies .~~ This combination of 5-FU and rIFN a-2a versus the same 5-FU regimen alone is being examined in Phase I11 trials of previously un- treated patients with advanced colorectal cancer to de-

termine the extent to which rIFN a-2a improves the results with this particular 5-FU regimen.

In a dose-escalated pilot study in patients with met- astatic gastrointestinal cancers,85 rIFN a-2a in doses ranging from 3 to 9 X lo6 U/m2/day for 7 days was combined with 5-FU 370-425 mg/m2 and high-dose leucovorin 500 mg/m2 administered by bolus for 5 con- secutive days per month. Among patients with ad- vanced colorectal carcinoma, an overall objective re- sponse rate of 44% was demonstrated. The toxicity of the addition of rIFN a-2a to 5-FU and high-dose leuco- vorin was examined in matched cycles (that is, the IFN a-2a was administered in the second cycle but not in the first) and was found to increase the incidence of mucositis from 0% to 31%, diarrhea from 0% to 23%, and decrease the median platelet nadir from 216-164 X 103/pL. The median leukocyte and granulocyte na- dirs were not affected. The toxicity induced by the ad- dition of rIFN a-2a occurred in a dose-dependent man- ner and prohibited the escalation of 5-FU dose. The authors recommended the 5-day regimen of 5-FU 370 mg/m2 and leucovorin 500 mg/m2 with rIFN a-2a 5 X lo6 U/mz/day as a regimen with acceptable toxicity that deserves additional study in a Phase I1 trial.

Because the cytotoxicity of 5-FU is partly related to its incorporation into RNA, natural nucleosides such as uridine compete with 5-FU for this incorporation. PALA decreases uridine and cytidine pools by inhibit- ing pyrimidine synthesis and thus enhances the activity of 5-FU.s6*87 Phase I trials of PALA established that at high doses, the dose-limiting toxicity was neurotoxicity, specifically cerebellar ataxia. However, it also has been shown that maximal inhibition of pyrimidine synthesis occurs at a much lower dose l e ~ e l . ~ ~ , ~ ~ In a Phase I1 study of weekly treatment with PALA at 250 mg/m2 and 5-FU 2600-3250 mg/m2 by 24-hour continuous infusion in advanced colorectal cancer, response rates as great as 43% were dem~nstrated.~’ The regimen in- corporating 5-FU 2600 mg/m2 was well-tolerated, with severe diarrhea and neurotoxicity noted in only a few patients. A second study used PALA 250 mg/m2 and dose-escalated bolus 5-FU at 600, 700, or 800 mg/m2 administered weekly, followed by a 2-week rest.” An overall response rate of 35% was seen. Significant diar- rhea was noted in 24% of patients at the 700 mg/m2 dose and 43% at the 800-mg/m2 dose. Significant he- matologic toxicity (Grade 4 leukopenia) occurred only at the 800-mg/mz dose. Phase 111 trials comparing weekly continuous infusion 5-FU and bolus PALA with other regimens of biochemically modulated 5-FU are being conducted by the Eastern Cooperative Oncology Group (ECOG) and the Southwest Oncology Group (SWOG) and are depicted in Table 4. These studies also

Nonsurgical Management/Vaughn and Huller 4287

directly compare the 5-FU/PALA regimen to the weekly infusional high-dose 5-FU regimen alone and should help define the importance of PALA in this reg- imen.

Methotrexate and cisplatin are other agents that have been used in combination with 5-FU. Methotrex- ate appears to enhance 5-FU cytotoxicity by inhibition of purine metabolism, resulting in an accumulation of phosphoribosylpyrophosphate and, subsequently, an increased formation of 5-FU/ribonucleotide com- p o u n d ~ . ~ ~ The sequential timing of administration of the two agents has been shown to be important. In a prospective randomized trial by Marsh et al.,92 5-FU was given 1 or 24 hours after the administration of methotrexate. A statistically improved response rate and survival benefit was demonstrated with the longer interval regimen. Multiple randomized studies have compared sequential methotrexate/5-FU combinations with 5-FU alone.73,77*93,94 Only one of these showed a statistically significant increase response rate for the methotrexate/5-FU regimen,93 whereas the other stud- ies showed roughly equivalent response rates. Two of the studies showed significant improvement in quality of life variables, such as weight gain, performance sta- tus, and relief of symptoms, compared with those asso- ciated with single-agent 5-FU the rap^.^^,^^ However, in one of these studies, the regimen was inferior to 5-FU/ low-dose leucovorin in terms of response rate and qual- ity of life variables.73 Thus, although methotrexate mod- ulates 5-FU and this combination may be modestly bet- ter than 5-FU alone, most investigators agree that it is not superior to 5-FU/leucovorin in overall cytotoxicity.

Cisplatin was demonstrated to be synergistic with 5-FU in the murine L1210 leukemia line.95 However, initial Phase I1 trials of bolus 5-FU given with cisplatin resulted in excessive myelosuppression, principally sig- nificant leukopenia and granul~cytopenia .~~,~~ Random- ized trials of bolus 5-FU/cisplatin have failed to show any benefit compared with 5-FU a l ~ n e . ~ ~ , ~ ' Thus, the addition of cisplatin to 5-FU appears to add significant toxicity without an appropriate increase in benefit and is not recommended as first-line therapy in metastatic colorectal cancer. There are reports that the addition of meth~trexate~~ or cisplatin"' to 5-FU may result in re- sponses in patients who have failed to respond to 5-FU alone, suggesting that these combinations may be use- ful as salvage regimens in selected patients.

An alternate strategy to biochemical modulation of bolus 5-FU is to enhance its cytotoxicity by alterations of dose and schedule, including continuous infusion. 5-FU has a short half-life (approximately 10-20 min- utes)"' and preclinical studies have addressed this as a basis for ineffective cytotoxicity.lo2 Dose escalation stud-

ies in humans have established 300 mg/m2/day as the maximum tolerated dose for protracted continuous in- fusion (10-week infusion). At this dose, an objective response rate of 38% was seen in patients with meta- static colorectal ~ a n c e r . ~ ' ~ , ~ ' ~ A Phase I11 trial comparing this dose and schedule of 5-FU with standard 5-day bolus 5-FU alone was performed by the Mid-Atlantic Oncology Program (MAOP).'05 A statistically signifi- cant improved response rate was seen in the continuous infusion treatment arm (30%) compared with the bolus treatment arm (7%; P < 0.001). Despite this improve- ment in response rate, overall median survival was identical in the two treatment arms.

Different patterns of toxicity were observed for each of these regimens. In the standard bolus treatment arm, bone marrow suppression, especially leukopenia, was seen in 14% of patients and stomatitis was noted in 13%. In the continuous infusion group, leukopenia was seen in only 1% of patients and stomatitis in 4%. How- ever, palmar-plantar erythrodysesthesia or "the hand- foot syndrome," a relatively uncommon side effect with bolus 5-FU therapy, occurred in 24% of patients who received protracted infusional therapy. lo6 This syndrome was never life-threatening and was fully re- versible with interruption of therapy. Biochemical mod- ulation of continuous infusion 5-FU with other agents is being studied. Bolus cisplatin has been combined with continuous infusion 5-FU and has been compared with continuous infusion 5-FU al~ne.~ '~,~ ' ' The objective of the combined continuous infusion 5-FU/cisplatin regi- men was to avoid the severe dose-limiting myelotoxic- ity seen in bolus 5-FU/cisplatin trials, while potentiat- ing 5-FU cytotoxicity. In a randomized study that com- pared bolus cisplatin with a 5-day continuous-infusion 5-FU regimen, an increased response rate was observed than that seen with 5-FU alone (25% versus 3%; P = 0.001), but survival was not affected, and toxicity was in~reased.''~ In the second trial, protracted infusion 5-FU 300 mg/m2/day was combined with bolus cispla- tin and compared with the infusional5-FU alone treat- ment.'" No benefit was demonstrated from the addi- tion of cisplatin to the regimen in terms of response rates (32% for 5-FU/cisplatin versus 27% for 5-FU). The addition of cisplatin to infusional5-FU can not be recommended.

Thus, significant progress has been made with re- spect to the administration of 5-FU and its modulators. However, questions remain about the optimum way to treat advanced colorectal cancer with 5-FU-based regi- mens, both in terms of the benefit of infusional versus bolus therapy and in the role of the various 5-FU modu- lators. Two ongoing national Phase I11 trials (Table 4) are studying these issues. ECOG 2290 compares weekly

4288 CANCER Supplement June 15, 1993, Volume 71, No. 12

Table 4. Ongoing Phase I11 Trials in Advanced Colorectal Cancer

ECOG 2290

Randomize 9 5-FU 2600 mg IV by continuous infusion over 24 h every wk

5-FU 2600 mg IV by continuous infusion over 24 hr and PALA 250 mg/m’ IV every wk

5-FU 600 mg/m’ IV and leucovorin 125 mg/m2 by mouth every wk x 6,2-week rest, repeat

5-FU 600 mg/m’ IV and leucovorin 500 mg/m’ IV every wk x 6,2-week rest, then repeat

5-FU 750 mg/m’ IV x 5 d then weekly and rIFNa-Za 9 million units 3 x/wk

SWOG 8905

Randomize 9 5-FU 500 mg/m2 IV q every d x 5 d every 5 wk 5-FU 425 mg/m2 IV + low-dose leucovorin 20

mg/m’ IV every d x 5 d every 4 wk x 2 then every 5 wk

5-FU 600 mg/m’ IV + high-dose leucovorin 500 mg/m’ IV on d 1,8,15, and 22 every 4 wk

5-FU 300 mg/m’ by continuous infusion IV d 1-28 every 5 wk

5-FU 200 mg/m’ by continuous infusion IV d 1-28 + low-dose leucovorin 20 mg/m’ on d 1,8,15, and 22 every 5 wk

5-FU 2600 mg/m2 by 24-hr continuous infusion IV d 1,8,15, and 22 every 4 wk

5-FU 2600 mg/m2 by 24-hr continuous infusion preceded by PALA 250 mg/m’ IV d 1,8,15, and 22 every 4 wk

5-FU: 5-fluorouracil; I V intravenous; PALA: phosphonacetyl-L-aspartate; rIFNa-Za: recombinant interferon alpha-2a.

high-dose 24-hour infusional 5-FU alone to this same regimen of 5-FU given with weekly PALA, to weekly bolus 5-FU with high-dose leucovorin (as per the GITSG study) and to weekly bolus 5-FU with weekly oral leucovorin or rIFN a-2a. SWOG 8905 compares a 5-day regimen of bolus 5-FU, to the NCCTG and GITSG regimens for bolus 5-FU/leucovorin, to pro- tracted continuous infusion 5-FU with or without low- dose weekly bolus leucovorin, to weekly high-dose in- fusional5-FU with or without weekly PALA. The goal of these intergroup trials with multiple treatment arms is to address the relative risks/benefits of these 5-FU- based regimens to define the optimal systemic chemo- therapy for patients with metastatic colorectal cancer.

A final issue of the treatment of advanced colorec- tal cancer with 5-FU-based chemotherapy that should be addressed is when to commence treatment. The Nordic Gastrointestinal Tumor Adjuvant Therapy Group recently reported their prospective trial in which a regimen of 5-FU, leucovorin, and methotrexate was administered to patients with metastatic colon cancer on diagnosis of advanced disease or development of

symptoms. Ninety-two patients received treatment on presentation, and 51 of 90 patients in the “expectancy” group received treatment. Overall survival was im- proved in the initially treated group compared with the expectancy group, with a difference in median survival of 5 months. The symptom-free period and the time to disease progression were longer in the initially treated group (median differences of 8 and 4 months, respec- tively). Thus, the authors concluded that early treat- ment of patients with asymptomatic advanced colorec- tal cancer prolongs survival, the asymptomatic period, and the time to disease progres~ion.’~~

Because most patients with advanced colorectal cancer eventually die of their disease despite chemo- therapy, novel therapeutic strategies are being devel- oped, including the use of monoclonal antibodies (MoAb), biologic response modifiers, and gene therapy. MoAb therapy for colorectal cancer has been re- viewed.”’ Among the most widely used murine MoAb are those directed against the tumor-associated anti- gens CEA, C017-1A, or TAG-72. The unconjugated murine 17-1A has been the most widely studied MoAb in advanced colorectal cancer. Several studies have demonstrated that this MoAb could be safely adminis- tered, but with objective partial response rates of less than 5% and with minor responses or stable disease in 25% of patient^."^-"^ A dose-response relationship with this agent has not been demonstrated. MoAb with specificity for tumor cells also offer the potential to de- liver conjugated cytotoxic moieties to the tumor with decreased exposure to normal tissues. Radionuclides are advantageous for use as the cytotoxic moiety be- cause internalization and activation are not needed for these agents to be effective, and sterilization of nonan- tigen-expressing neighboring cells may be a~hieved.”~

At the University of Pennsylvania, the murine MoAb B72.3 conjugated to 90Y has been administered to patients with refractory TAG-72 expressing tumors, including colorectal cancer. In a Phase I trial of this MoAb in 25 patients, reversible myelosuppression, principally thrombocytopenia, was the dose-limiting toxicity. Doses as large as 15 mCi of 90Y were adminis- tered with no objective responses dem~nstrated.”~ In an effort to increase the amount of radioisotope admin- istered, a second Phase I pilot study was performed using the MoAb in combination with ethylenediamine tetraacetic acid (EDTA), a metal chelator that increases urinary clearance of 90Y.116 In this study, increased doses of the radioisotope have been administered, with dose-limiting reversible thrombocytopenia occurring at 15 mCi/m2. No objective responses have been seen in this heterogeneous group of mostly pretreated patients. Future studies may attempt to increase the efficacy of radiolabeled MoAb B72.3 therapy by multiple dosing

Nonsurgical Management/Vaughn and Haller 4289

and by dose escalation of the agent in combination with the radioprotectant interleukin-1 (IL-1), a strategy that has proven successful in murine models.”’,”* Based on the synergistic effects of 5-FU and radiation demon- strated preclinically by By field et al.,lI9 the radiolabeled MoAb B72.3 may additionally be tested in combination with protracted infusional 5-FU in an attempt to attain chemosensitization of the tumor cells to the radionu- clide.

Among the immunobiologic agents that have been used in metastatic colorectal cancer, rIFN a-2a has been studied most extensively. However, this agent’s pri- mary role is as a 5-FU modulator, not as an immuno- modulatory agent.’20 In advanced colorectal cancer, re- combinant IL-2, a T-cell growth factor that stimulates proliferation of lymphokine-activated killer cells, has been used. Trials of IL-2 alone, IL-2 plus lymphokine- activated killer cells, and IL-2 with rIFN a-2a in patients with advanced colorectal cancer have demonstrated re- sponse rates of O%, 13%, and 7%, respectively.”’ Toxic- ity associated with the administration of IL-2 has been significant. Finally, an emerging field in the treatment of solid tumors that has been pioneered by Rosenberg et

at the National Cancer Institute is that of gene a1.121,122

therapy. Using the administration of genetically modi- fied tumor-infiltrating lymphocytes and IL-2, this ther- apy has been piloted in patients with advanced mela- noma. Preliminary trials are underway at the National Cancer Institute using this treatment in patients with advanced colorectal cancer (personal communication, Rosenberg SA).

Summary

Ongoing research into the development of innovative therapies such as those discussed here and additional investigation of the optimum use of 5-FU will continue to examine the role of treatment in providing improved quality of life and prolonged survival of patients with recurrence of colorectal carcinoma. Optimum manage- ment will continue to be dependent on a multidisciplin- ary approach that uses the various modalities provided by surgery, radiation oncology, and medical oncology. These advances, as measured by their success in the management of recurrent colorectal cancer, may prove most useful in the adjuvant treatment of early stage tumors.

References

1. Boring CC, Squires TS, Tong T. Cancer statistics 1991. CA 1991;

2. Mayer RJ, O’Connell MJ, Tepper JE, Wolmark N. Status of adju- vant treatment of colorectal cancer. J Natl Cancer lnst 1989;

41 19-36.

81: 1359-64.

3. Moertel CG, Fleming TP, Macdonald JS, Haller DG, Laurie JA, Goodman PJ, et al. Levamisole and fluorourad for adjuvant treat- ment of resected colon cancer. N Engl J Med 1990; 322:352-8.

4. Moertel CG, Fleming T, Macdonald J, Haller D, Laurie J . The intergroup study of fluorouracil(5-FU) plus levamisole (lev) and levamisole alone as adjuvant therapy for stage C colon cancer. Proc Am Soc Clin Oncol 1992; 11:161.

5. Gastrointestinal Tumor Study Group. Prolongation of disease- free interval in surgically treated rectal cancer. N Engl ] Med

6. Gastrointestinal Tumor Study Group, Douglass HO Jr., Moertel CG, Mayer RJ, Thomas PRM, Lindbland AS, et al. Survival after post-operative combination treatment of rectal cancer. N Engl I Med 1986; 315:1294-5.

7. Krook JE, Moertel CG, Gunderson LL, Wieand HS, Collins RT, Beart RW, et al. Effective surgical adjuvant therapy for high risk rectal cancer. N Engl J Med 1991; 324:709-15.

8. Olson RM, Perencevich NP, Malcolm AW, Chaffey JT, Wilson RE. Patterns of recurrence following curative resection of adeno- carcinoma of the colon and rectum. Cancer 1980; 45:2969-74.

9. Rich T, Gunderson LL, Lew R, Galdabini J, Cohen AM, Donald- son G. Patterns of recurrence of rectal cancer after potentially curative surgery. Cancer 1983; 521317-29,

10. Schilsky RL, Brachman DG. Adjuvant chemotherapy and radia- tion therapy in colorectal cancer. In: DeVita VJ, Hellman S, Ro- senberg SA, editors. PPO Updafes of Principles and Practice of Oncology 1992; 6:l-12.

11. Wollmark N, Fisher B. An analysis of survival and treatment failure following abdominoperitoneal and sphincter-saving re- section in Dukes’ B and C rectal carcinoma. Ann Surg 1986;

12. Cohen AM, Minsky BD. Aggressive surgical management of lo- cally advanced primary and recurrent rectal cancer. Dis Colon Rectum 1990; 33:432-8.

13. Beart RW, Martin JK, Gunderson LL. Management of recurrent rectal cancer. Mayo Clin Proc 1986; 61:448-50.

14. Schiessel R, Wunderlich M, Herbst F. Local recurrence of colorec- tal cancer: effect of early detection and aggressive surgery. Br J Surg 1986; 73:342-4.

15. Dobrowsky W, Schmid AP. Radiotherapy of presacral recur- rences following radical surgery of rectal cancer. Dis Colon Rec- tum 1985; 28917-9.

16. Pacini P, Cionini L, Pirtoll L, Ciatto S, Tucci E, Sebaste L, et al. Symptomatic recurrence of carcinoma of the rectum and sig- moid: the influence of radiotherapy on the quality of life. Dis Colon Rectum 1986; 29365-8.

17. Rominger CJ, Gelber RD, Gunderson LL, Conner N. Radiation therapy alone or in combination with chemotherapy in the treatment of residual or inoperable carcinoma of the rectum and rectosigmoid or pelvic recurrence following colorectal surgery: Radiation Therapy Oncology Group Study (76-16). Am J Clin oncor 1985; 8:118-27.

18. Danjoux CE, Gelber RD, Catton GE, Klaassen DJ. Combination chemotherapy for residual, recurrent, or inoperable carcinoma of the rectum: ECOG Study (EST 3276). Int J Radiat Oncol Biol

19. Dobrowsky W. Mitomycin C, 5-fluorouracil, and radiation in advanced, locally recurrent rectal cancer. Br ] Radiol 1992;

20. Gunderson LL, Martin JK, O’Connell MJ, Beart RW, Kvols LK, Nagorney DM. Residual, recurrent, or unresectable gastrointesti- nal cancer. Cancer 1985; 55:2250-8.

21. Patt YZ, Peters RE, Chuang VP, Wallace S, Claghorn L, Mavligit

1985; 312:1465-72.

204:480-Y,

Phys 1985; 111765-71.

65:143-7.

4290 CANCER Supplement June 25, 2993, Volume 71, No. 12

G. Palliation of pelvic recurrence of colorectal cancer with intra- arterial 5-FU and mitomycin. Cancer 1985; 56:2175-80.

22. Patt YZ, Mavligit GM. Arterial chemotherapy in the manage- ment of colorectal cancer: an overview. Semin Oncol 1991;

23. Meijer S, de Rooij I‘D, Derksen EJ, Boutkan H, Cuesta MA. Cryosurgery for locally recurrent rectal cancer. Eur ] Surg Oncol

24. Bengmark 5, Hafstrem L. The natural history of primary and secondary malignant tumors of the liver. Cancer 1969; 23:198- 202.

25. Willet CG, Tepper JE, Cohen AM, Orlow E, Welch CE. Failure patterns following curative resection of colonic carcinoma. Ann Surg 1984; 200:685-90.

26. Haller DG. Prognostic factors for liver metastases from colorec- tal cancer. In: Mastromarino A, editor. The biology and treat- ment of colorectal cancer metastases. Boston: Martinus Nijhoff,

27. Steele G, Ravikumar TS. Resection of hepatic metastases from colorectal cancer: biological perspectives. Ann Surg 1989;

28. Steele G, Bleday R, Mayer RJ, Lindblad A, Petrelli N, Weaver D. A prospective evaluation of hepatic resection for colorectal me- tastases to the liver: Gastrointestinal Tumor Study Group Proto- col 6584. ] Clin Oncol 1991; 9:1105-12.

29. Einhorn LH. Improvements in fluorouracil chemotherapy? \ Clin Oncol 1989; 7:1377-9.

30. Collins JM. Pharmacologic rationale for regional drug delivery. J C h i Oncol 1984; 2:498-504.

31. Breeedis C, Young C. The blood supply of neoplasms in the liver. A m PatRol 1954; 30:969-85.

32. Chen GHG, Gross JF. Intra-arterial infusion of anticancer drugs: theoretical aspects of drug delivery and review of responses. Cancer Treatment Rep 1980; 64:31-40.

33. Ensminger WD, Rosowsky A, Raso V, Levin DC, Glode M, Come 5, et al. A clinical pharmacological evaluation of hepatic arterial infusion of 5-fluoro-2’-deoxyuridine and 5-fluorouracil. Cancer Res 1978; 38:3784-92.

34. Tandon RN, Bunnell IL, Copper RG. The treatment of metastatic carcinoma of the liver by percutaneous selective hepatic artery infusion of 5-fluorouracil. Surgery 1973; 73:118.

35. Ansfield FJ, Ramirez G, Davis HL Jr., Wirtenanen GW, Johnson RO, Bryan GT, et al. Further clinical studies with intrahepatic arterial infusion with 5-fluorouracil. Cancer 1975; 36:2413-7.

36. Oberfield RA, McCaffrey JA, Polio J, Clouse ME, Hamilton T. Prolonged and continuous infusion chemotherapy in advanced metastatic liver adenocarcinoma from colorectal primary. Cancer

37. Watkins E Jr., Khazei AM, Nahra KS. Surgical basis for arterial infusion chemotherapy of disseminated carcinoma of the liver. Surg Gynecol Obstet 1970; 130:581-605.

38. Cady B, Oberfield RA. Regional infusion of hepatic metastases from carcinoma of the colon. A m ] Surg 1974; 127:220-7.

39. Smiley 5, Schouten J, Chang A, Ramjrez G. Intrahepatic arterial infusion with 5-FU for liver metastases of colorectal carcinoma [abstract]. Proc A m Soc Clin Oncol 1981; 22:391.

40. Kemeny N, Sugarbaker PH. Treatment of metastatic cancer to liver. In: DeVita VJ, Hellman 5, Rosenberg SA, editors. Princi- ples and practice of oncology. 3rd ed. Philadelphia: JB Lippin-

41. Buchwald H, Grage TB, Vassilopoulous PP, Rohde TD, Varco RL, Blackshear PJ. Intraarterial infusion chemotherapy for he- patic carcinoma using a totally implantable infusion pump. Cancer 1980; 455366-9.

42. Patt YZ, Boddie A, Soski M, et al. Arterial therapy: percutaneous

5:478-90.

1992; 18:255-7.

1986:253-6.

210:127-38.

1979; 44~414-23.

cott CO., 1989:2275-98.

catheters or implantable infusion devices (meditronics DADS, Infusaid). In: Ensminger WD, Selam JL, editors. Infusion sys- tems in medicine. Mount Kisco, NY: Futura Publishing Co,

43. Kemeny V, Daly J, Reichman B, Geller N, Botet J, Oderman P. Intrahepatic or systemic infusion of fluordeoxyuridine in pa- tients with liver metastases from colorectal carcinoma. Ann In- tern Med 1987; 107:459-65.

44. Chang AE, Schneider PD, Sugarbaker PH, Simpson C, Culnare M, Steinberg SM. A prospective randomized trial of regional versus systemic continuous 5-fluorodeoxyuridine chemother- apy in the treatment of colorectal liver metastases. Ann Surg

45. Hohn DC, Stagg RJ, Friedman MA, Hannigan JF, Rayner A, Ignoffo RJ. A randomized trial of continuous intravenous versus hepatic intraarterial floxuridine in patients with colorectal cancer metastatic to the liver: The Northern California Oncology Group Trial. 1 Clin Oncol 1989; 7:1646-54.

46. Martin JK, O’Connell MJ, Wieand HS, Fitzgibbon RJ Jr., Mail- liard JA, Rubin J, et al. Intraarterial floxuridine versus systemic fluorouracil for hepatic metastases for colorectal cancer: a ran- domized trial. Arch Surg 1990; 125:1022-7.

47. Rougier P, Laplanche A, Haguier M, Hay J, Ollivier J, Escat J, et al. Hepatic arterial infusion of floxuridine in patients with liver metastases from colorectal carcinoma: long term results of a pro- spective randomized trial. \ Clin Oncol 1992; 10:1112-8.

48. Kemeny N, Conti J, Seiter K. A Phase I trial of intrahepatic fluo- rodeoxyuridine and systemic 5-fluorouracil t leucovorin in met- astatic colorectal cancer [abstract]. Proc An1 Soc Clin Oncol 1992; 11:194.

49. Safi F. Continuous simultaneous intraarterial (IA) and intrave- nous (IV) therapy of liver metastases of colorectal carcinoma: results of a prospective randomized trial [abstract]. Proc An1 Soc Clin Oncol 1992; 11:169.

50. Kemeny N. Review of regional therapy of liver metastases in colorectal cancer. Senrin Oncol 1992; 19:155-162.

51. Stagg RJ, Venook AP, Chase JL, Lewis BJ, Warren RS, Roh M, et al. Alternating hepatic intraarterial FUdR and 5-FU: a less toxic regimen for treatment of liver metastases from colorectal cancer. 1 Nafl Cancer Znst 1991; 8:423-8.

52. Seifer S, Kemeny N, Berger M, Niedzwiecki D, Chapman D, Sigurdson E, et al. A randomized trial of intrahepatic infusion of FUdR with dexamethasone versus FUdR alone in the treatment of metastatic colorectal cancer [abstract]. Proc A m Soc Clin Oncol 1990; 9:lOB.

53. Von Roemling R, Hrushsky WJ. Determination of therapeutic index of fluoxuridine by its circadian infusion pattern. ] Natl Cancer Znst 1990; 82:386-93.

54. Hohn DC, Stagg RJ, Price DC, Lewis BJ. Avoidance of gastro- duodenal toxicity in patients receiving hepatic arterial 5-flUOrO- 2’-deoxyuridine. \ Clin Oncol 1985; 3:1257-60.

55. Patt YZ, Mavligit GM, Chuang VP, Wallace S, Johnston 5, Ben- jamin RS, et al. Percutaneous hepatic arterial (HAI) of mitomy- cin-C and floxuridine: an effective treatment for metastatic colo- rectal carcinoma in the liver. Cancer 1980; 46261-5.

56. Patt YZ, Boddie AW, Charnsangavej C, Ajani JA, Wallace 5, Soski M. Hepatic arterial infusion with floxuridine and cisplati- num: overriding importance of antitumor effect versus degree of tumor burden as determinants of survival among patients with colorectal cancer. J Clin Oncol 1986; 4:1356-64.

57. Patt YZ, Peters R, Chuang VP, Wallace 5, Mavligit G. Effective retreatment of patients with colorectal cancer and liver metas- tases. A m \ Med 1983; 75:237-40.

58. Patt YZ, Roh M, Chase J, Levin B, Hohn D. A Phase I trial of hepatic arterial infusion of floxuridine and folinic acid for colo-

1988~189-99.

1987; 206:685-93.

Nonsurgical Management/Vaughn and Huller 4291

rectal cancer metastatic to the liver [abstract]. Proc A m Soc Clin Oncol 1990; 9:118.

59. Kemeny N, Cohen A, Bertino JR, Sigurdson ER, Botet J, Oder- man P. Continuous infusion of floxuridine and leucovorin through an implantable pump for the treatment of hepatic me- tastases from colorectal carcinoma. Cancer 1989; 659446-50.

60. Mavligit GM, Zukwiski AA, Cutterman JU, Salem P, Charnsan- gavej C, Wallace S. Splenic versus hepatic artery infusion of interleukin-2 in patients with liver metastases. Clin Oncoll990;

61. Mavligit GM, Zulewski A, Wallace S. Tumor regression after hepatic arterial infusion of recombinant tumor necrosis factor in patients with colon cancer metastatic to the liver [abstract]. Proc A m Soc Clin Oncol 1990; 9:118.

62. Wallace S, Carrasco C , Ajani J, et al. Embolization of neuroendo- crine hepatic metastases. In: Levin B, editor. Gastrointestinal cancer: current approaches to diagnosis and treatment. Austin: University of Texas Press, 1988:337-49.

63. Venook AP, Stagg RJ, Lewis BJ. Chemoembolization for hepato- cellular carcinoma. Clin Oncol 1990; 8:1108-14.

64. Hunt T, Flowerdew A, Birch S, Williams JP, Mullae MA, Taylor I. Prospective randomized controlled trial of hepatic arterial em- bolization or infusion chemotherapy with 5-fluorouracil and de- gradable starch microspheres for colorectal liver metastases. Br ] Surg 1990; 77:779-82.

65. Wallace S, Carrasco CH, Charnsangavej C, Richli WR, Wright K, Gianturco C . Hepatic artery infusion and chemoembolization in the management of liver metastases. Cardiovasc Intervent Radiol

66. Daniels JR, Pentocost MJ, Teitelbaum G, Sternlicht M. Hepatic artery chemoembolization (HAE) for carcinoma of the colon us- ing with Angiostat” collagen, cisplatin, doxorubicin, and mito- mycin: response, survival, and serum drug level [abstract]. Proc A m Soc Clin Oncol 1992; 11:171.

67. Livraghi T, Claudio V, Lazzaroni S. Liver metastases: results of percutaneous ethanol injection in 14 patients. Radiology 1990;

68. Atkinson D, Zemel R, Weaver ML, Onik G . Hepatic cryosurgery for metastatic carcinoma [abstract]. Proc Am Soc Clin Oncoll992; 11:165.

69. Abbruzzese JL, Levin B. Treatment of advanced colon cancer. Hematol Oncol Clin North Am 1989; 3:135-53.

70. Arbuck SG. 5-FU/leucovorin: biochemical modulation that works? Oncology 1987; 1:61-71.

71. Petrelli N, Herrera L, Rustum Y, Burke P, Creaven P, Stulc J, et al. A prospective randomized trial of 5-fluorouracil versus 5-flu- orouracil and high dose leucovorin versus 5-fluorouracil and methotrexate in previously untreated patients with advanced colorectal carcinoma. ] Clin Oncol 1987; 5:1559-65.

72. Petrelli N, Douglass HO Jr,, Herrera L, Russell D, Stablein DM, Bruckner HW, et a]. The modulation of fluorouracil with leuco- vorin in metastatic colorectal carcinoma: a prospective random- ized phase 111 trial. ] Clin Oncol 1989; 7:1419-26.

73. Poon MA, OConnell MJ, Moertel CG, Wieand HS, Cullinan SA, Everson LK, et al. Biochemical modulation of fluorouracil: evi- dence of significant improvement of survival and quality of life in patients with advanced colorectal carcinoma. ] Clin Oncol

74. Poon MA, O’Connell MI, Wieand HS, Krook JE, Gerstner JB, Tschetter LK, et al. Biochemical modulation of fluorouracil with leucovorin: confirmatory evidence of improved therapeutic eHi- cacy in advanced colorectal cancer. ] Clin Oncol 1991; 9:1967- 72.

75. Doroshow JH, Mdthauf P, Leong L, Margolin K, Litchfield T, Akman S, et al. Prospective randomized comparison of fluoro-

8:319-24.

1990; 13: 153-60.

179:709-12.

1989; 7:1407-18.

uracil versus fluorouracil and high dose continuous infusion leucovorin for the treatment of advanced colorectal cancer in patients previously unexposed to chemotherapy. J Clin Oncol

76. Ehrlichman C, Fine S, Wong A, Elhakim T. A randomized trial of fluorouracil and folinic acid in patients with metastatic colo- rectal cancer. 1 Clin Oncol 1988; 6:469-75.

77. Valone FH, Friedman MA, Wittlinger PS, Drakes T, Eisenberg PD, Malec M, et al. Treatment of patients with advanced colorec- tal carcinomas with fluorouracil alone, high-dose leucovorin plus fluorouracil, or sequential methotrexate, fluorouracil, and leucovorin: a randomized trial of the Northern California Oncol- ogy Group. ] Clin Oncol 1989; 7:1427-36.

78. Gerstner J, OConnell MJ, Wieand HS, Buroker TR, Krook J. A prospective randomized clinical trial comparing 5-FU combined with either high- or low-dose leucovorin for the treatment of advanced colorectal cancer [abstract]. Proc A m Soc Clin Oncol 1991; 10:134.

79. Advanced Colorectal Cancer Meta-Analysis Project. Modula- tion of 5-fluorouracil in patients with advanced colorectal cancer: evidence in terms of response rate. 1 Clin Oncol 1992;

80. Gewert DR, Shah S, Clemens MJ. Inhibition of cell division by interferons: changes in the transport and intracellular metabo- lism of thymidine in human lymphoblastoid (Daudi) cells. Eur ] Biochem 1981; 116:487-92.

81. Elias L, Crissman HA. Interferon effects on the adenocarcinoma 38 and HL-60 cell lines: antiproliferative responses and syner- gistic interactions with halogenated pyrimidine antimetabolites. Cancer Res 1988; 48:4868-73.

82. Elias L, Sandoval JM. Interferon effects upon fluorouracil metab- olism by HL-60 cells. Biochem Biophys Res Commun 1989;

83. Wadler SW, Schwartz EL, Goldmann M, Lyver A, Rader M, Zimmerman M, et al. Therapy with 5-fluorouracil and recombi- nant alpha-2a-interferon: an active regimen against advanced colorectal carcinoma. ] Clin Oncol 1989; 7:1769-75.

84. Pazdur R, Ajani JA, Patt YZ, Winn R, Jackson D, Shepard B, et al. Phase I1 study of fluorouracil and recombinant interferon alfa-2a in previously untreated advanced colorectal carcinoma. ]

85. Grem JL, McAtee N, Murphy RF, Balis FM, Steinberg SM, Ha- milton JM, et al. A pilot study of interferon alfa-2a in combina- tion with 5-fluorouracil and high dose leucovorin in metastatic gastrointestinal cancer. Clin Oncol 1991; 9:1811-20.

86. Spiegelman S, Sawyer R, Nayak R, Ritzi R, Stolfi R, Martin D. Improving the anti-tumor activity of 5-fluorouracil by increas- ing its incorporation into RNA by metabolic modulation. Proc Natl Acad Sci USA 1980; 77:4966-70.

87. Grem JL, King SA, O’Dwyer PJ, Leylan-Jones B. Biochemistry and clinical activity of N- (phosphonacety1)-L-aspartate: a re- view. Cancer Res 1988; 48:4441-54.

88. Casper ES, Vale K, Williams LJ, Martin DS, Young CW. Phase 1 and clinical pharmacological evaluation of biochemical modula- tion of 5-fluorouracil with N-(phosphonacety1)-L-aspartatic acid. Cancer Res 1983; 43:2324-8.

89. O’Dwyer PJ, Paul AR, Walczak J, Weiner LM, Litwin S, Comis RL. Phase 11 study of biochemical modulation of fluorouracil by low-dose PALA in patients with colorectal cancer. ] Clin Oncol

90. Kemeny N, Conti J, Seiter K, Niedzwiecki D, Botet J, Martin D, et al. Biochemical modulation of bolus fluorouracil by PALA in patients with advanced colorectal cancer. ] Clin Oncol 1992;

91. Cadman E, Heimer R, Benz C. The influence of methotrexate

1990; 8~491-501.

10~896-903.

1631867-74.

crin oncor 1990; 8:2027-31.

1990; 9:1497-503.

10~747-52.

4292 CANCER Supplement June 25, 2993, Volume 71, No. 12

pretreatment on 5-fluorouracil metabolism in L1210 cells. Biol Chem 1981; 256:1695-704.

92. Marsh JC, Bertino JR, Katz KH, Davis CA, Durivage HJ, Rome LS, et al. The influence of drug interval on the effect of metho- trexate and fluorouracil in the treatment of advanced colorectal cancer. J C h Oncol 1991; 9:371-80.

93. Machiavelli M, Leone B, Romero A, Rabinovich MM, Vallejo CCC, Bianco A, et al. Advanced colorectal carcinoma: a prospec- tive randomized trial of sequential methotrexate + 5-fluoroura- cil vs. 5FU alone [abstract]. Proc A m Soc Clin Oncol 1990; 9:102.

94. The Nordic Gastrointestinal Tumor Adjuvant Therapy Group. Superiority of sequential methotrexate, fluorouracil, and leuco- vorin to fluorouracil alone in advanced asymptomatic colorectal carcinoma: a randomized trial. J Clin Oncol 1989; 7:1437-46.

95. Schabel FM Jr., Trade MW, Laster WR, Corbett TH, Griswold DP Jr. Cis-dichlorodiamine-platinum (11): combination chemo- therapy and cross-resistance studies with tumors of mice. Cancer Treat Rep 1979; 63:1459-73.

96. Whitehead RP, Fleming T, Macdonald JS, Ahmann F, Inamasu M. A Phase I1 study of 5-fluorouracil plus cisplatin for meta- static colorectal adenocarcinoma: a Southwest Oncology Group (SWOG) Study [abstract]. Proc Am Soc Clin Oncol 1989; 8:128.

97. O'Connell M, Moertel C, Kvols L, et al. A Phase 11 trial of inten- sive course 5-fluorouracil (5-FU) plus high dose cis-platinum (CDDP) in advanced colorectal cancer [abstract]. Proc A m Assoc Cancer Res 1985; 26:166.

98. Loehrer PJ Sr., Turner S, Kubilis l', Hui S, Correa J, Ansari R, et al. A prospective randomized trial of fluorouracil versus fluoro- uracil plus cisplatin in the treatment of metastatic colorectal cancer: a Hoosier Oncology Group trial. J Clin Oncol 1988;

99. Bruckner HW, Motwani BT. Chemotherapy of advanced cancer of the colon and rectum. Semin Oncol 1991; 18:443-61.

100. Petrelli NJ, Madejewicz S, Rustum Y, Herrera L, Craven PJ, Plager J, et al. Combination chemotherapy of cisplatinum and 5-fluorouracil for advanced colorectal adenocarcinoma. Cancer Chemother Pharmacol 1989; 23:57-60.

101. MacMillan WE, Wobery WH, Welling PG. Pharmacokinetics of 5-fluorourad in humans. Cancer Res 1978; 38:3479-82.

102. Drewinko 8, Yang LY. Cellular basis for the inefficacy of 5-FU in human colon carcinoma. Cancer Treat Rep 1985; 69:1391-8.

103. Lokich JJ, Bothe A, Fine N, Pem J. Phase I study of protracted venous infusion of 5-fluorouracil. Cancer 1981; 48:2565-8.

104. Lokich JJ. Colon and rectal cancer. In: Lokich JJ, editor. Cancer chemotherapy by infusion. Chicago: Precept, 1990:323-39.

105. Lokich JJ, Ahlgren JD, Gullo JJ, Philips JA, Fryer JG. A prospec- tive randomized comparison of continuous infusion fluorouracil with a conventional bolus schedule in metastatic colorectal carci- noma: a Mid-Atlantic Oncology Program Study. Clin Oncol

106. Lokich JJ, Moore C. Chemotherapy associated palmar-plantar erythrodysesthesia syndrome. Ann lntern Med 1984; 101:798- 800.

107. Hansen R, Ryan L, Anderson T, Quebbeman E, Haller D. A Phase 111 trial of bolus 5FU versus protracted infusion 5FU +/- cisplatin in metastatic colon cancer: An Eastern Cooperative On-

6~642-8.

1989; 7425-32.

cology Group study (EST 2286) [abstract]. Proc A m Soc Clin On- col 1992; 11:171.

108. Kemeny N, Israel K, Niedzwiecki D, Chapman D, Botet J, Minsky B, et al. Randomized study of continuous infusion fluo- rouracil versus fluorouracil plus cisplatinum in patients with metastatic colorectal cancer. J Clin Oncol 1990; 8:313-8.

109. The Nordic Gastrointestinal Tumor Adjuvant Therapy Group. Expectancy or primary chemotherapy in patients with advanced asymptomatic colorectal cancer: a randomized trial. J Clin Oncol

110. Mellstedt H, Frodin JE, Masucci G, Ragnhammar P, Fagerberg J, Hjelm A, et al. The therapeutic use of monoclonal antibodies in colorectal carcinoma. Semin Oncol 1991; 18:462-77.

11 1. Sears HF, Herlyn D, Steplewski Z, Koprowski H. Initial trial of a murine monoclonal antibody as an immunotherapeutic agent for gastrointestinal adenocarcinoma. Hybridoma 1986; 5:109- 123.

112. LoBuglio AF, Saleh MN, Lee J, Khazaeli MB, Carrano R, Holden H, et al. Phase I trial of multiple large doses of a murine monoclo- nal antibody C017-1A I. clinical aspects. J Natl Cancer lnsf

113. Sears HF, Herlyn D, Steplewski 2, Koprowski H. Phase I1 clini- cal trial of a murine monoclonal antibody cytotoxic for gastroin- testinal adenocarcinoma. Cancer Res 1985; 45:5910-3.

114. Knapp RC, Lau CC. Monoclonal antibodies and radioimmuno- conjugates: keeping feasibility at center stage. J Clin Oncol 1990;

115. Haller D, Cannon L, Keenan R, Maguire R. A Phase I trial of "Y labeled monoclonal antibody (MoAb) 872.3 (CYT-103-90Y) in refractory adenocarcinomas [abstract]. Antibody lmmunocon Ra- diopharm 1991; 4:202.

116. Haller D, Cannon L, Alavi A, Gulf0 J. A Phase I trial of 9 labeled monoclonal antibody (MoAb) B72.3 (CYT-103-90Y) ad- ministered with concurrent EDTA in refractory adenocarcinoma [abstract]. Antibody lmmunocon Radiopharm 1992: 5:147.

117. DeNardo SJ, DeNardo GL, Richman CM. Enhancement of clini- cal radioimmunotherapy for cancer and the role of cytokines. J Nut1 Cancer lnst 1992; 84:374-5.

118. Blumenthal R, Sharkey R, Goldenberg D. Dose escalation of radioantibody in a mouse model with the use of recombinant human interleukin-1 and granulocyte-macrophage colony-stim- dating factor intervention to reduce myelosuppression. J Natl Cancer Znst 1992; 84:399-407.

119. Byfield J, Calbro-Jones P, Klisak I, Kulhanian F. Pharmacologic requirements for obtaining sensitization of human tumor cells in vitro to combined 5-fluorouracil or ftorafur and x-rays. lnt J Radiat Oncol Biol Phys 1982; 8:1923-33.

120. Wadler S. The role of immunotherapy in colorectal cancer. Se- min Oncol 1991; 18:27-38.

121. Rosenberg S. The immunotherapy and gene therapy of cancer. J Clin Oncol 1992; 10:180-99.

122. Rosenberg SA, Aebersold P, Cornetta K, Kasid A, Morgan RA, Moen R, et al. Gene transfer into humans-immunotherapy of patients with advanced melanoma using tumor-infiltrating lym- phocytes modified by retroviral gene transduction. N Engl J Med

1992; 10:904-11.

1988; 80:932-6.

8:1938-40.

1990; 323~570-8.