• Non ci sono risultati.

Chemotherapy of Upper GI Neoplasms:Proven/Unproven 26

N/A
N/A
Protected

Academic year: 2022

Condividi "Chemotherapy of Upper GI Neoplasms:Proven/Unproven 26"

Copied!
10
0
0

Testo completo

(1)

5 6 7 8 9 1011 1 2 3 4 5 6 7 8 9 2011 1 2 3 4 5 6 7 8 9 3011 1 2 3 4 5 6 7 8 9 4011 1 2 3 4 5 6 7 8 9 5011 1 2 311

Chemotherapy of Upper GI Neoplasms:

Proven/Unproven

Niall C. Tebbutt and David Cunningham

Aims

To establish the role of chemotherapy for gastro-oesophageal tumours using cur- rently available evidence.

Introduction

In solid tumours, chemotherapy is frequently used both for palliation of advanced disease and also as an adjuvant therapy in order to improve the results of surgery in those patients with operable early stage disease. This chapter dis- cusses the role of chemotherapy in upper gas- trointestinal tract neoplasms, which comprise gastric and oesophageal cancers.

Advanced Disease

In the Western world, the majority of cases of oesophagogastric cancer present with advanced stage disease. Whilst loco-regional therapies such as surgery are the only treatments capable of achieving cure in early stage disease, they have an extremely limited role in advanced stage disease. Chemotherapy is used to control symptoms, maintain quality of life and prolong survival. Since advanced oesophagogastric cancer is incurable, it was important to ascer- tain the benefit attributable to the use of chemotherapy in this setting. Table 26.1 sum-

marises the results of trials comparing combi- nation chemotherapy regimens with best sup- portive care. Although these trials incorporate small numbers of patients, they have shown that chemotherapy achieves a statistically significant prolongation in median survival. It has also been demonstrated that the quality of life is also significantly improved in patients receiving chemotherapy, and financial analyses have shown that attainment of these benefits is cost effective [1]. It is now, therefore, accepted that patients with advanced oesophagogastric cancer who are sufficiently fit to receive chemotherapy should be considered for this treatment.

Single Agent Chemotherapy

A variety of chemotherapy agents have been evaluated using phase II studies in advanced

Table 26.1. Randomised trials comparing combination chemotherapy with best supportive case (BSC) in patients with advanced gastric cancer

Regimen Number of Median survival p patients (months)

ELF 10 10 <0.02

BSC 8 4

FAMTX 30 10 <0.001

BSC 10 3

FEMTX 17 12 <0.001

BSC 19 3

ELF, epirubicin, leucovorin, 5FU; FAMTX, 5FU, adriamycin, methotrexate; FEMTX, 5FU, epirubicin, methotrexate.

(2)

oesophagogastric cancer [2]. Results achieved in these studies are shown in Table 26.2.

Most studies include relatively small numbers of selected patients. Whilst it is difficult to compare the results between studies, it does provide an indication of which agents may have the greatest activity. The results demonstrate that relatively few agents can achieve response rates of greater than 25% in previously untreated patients.

Combination Chemotherapy Regimens

In an attempt to improve the results of chemotherapy, combination regimens consist- ing of those agents with the highest activity have been devised. Fluorouracil (5FU) has been a key component of many of these regimens because of its activity and its modest toxicity profile par- ticularly when administered by infusion sched- ules. An early combination regimen (FAM) consisted of 5FU, adriamycin and mitomycin-C (MMC). This combination regimen was estab- lished based on impressive response rates in phase II studies. The activity of this combina- tion regimen was confirmed in a randomised phase III study comparing treatment using FAM with adriamycin and MMC and 5FU, adri- amycin and methyl-CCNU in patients with advanced gastric cancer [3]. The FAM combi- nation regimen achieved the highest response rates (39%) and was also associated with the longest median survival (7.5 months).

Further chemotherapy regimens were devised in an attempt to improve on the FAM regimen.

The FAMTX regimen incorporated high dose methotrexate instead of MMC. A randomised study showed that FAMTX achieved superior response rates (41% versus 9%; p = 0.0001) and more prolonged median survival times (9.7 months versus 6.7 months; p = 0.004) than FAM [4]. More recently, two other combination regimens have been found to be equivalent to FAMTX in advanced gastric cancer. The response rates achieved using etoposide, leu- covorin and bolus 5FU (ELF), infused 5FU and cisplatin (FP) and FAMTX were 9%, 20% and 12% respectively and were not significantly different. The median survival times were 7.2 months with ELF, 7.2 months with FP and 6.7 months with FAMTX [5]. The authors con- cluded that all of these regimens had modest activity and that new strategies for advanced disease should be considered.

The combination of epirubicin 50 mg/m

2

, cisplatin 60 mg/m

2

and protracted venous infu- sion 5FU 300 mg/m

2

/day was developed at the Royal Marsden Hospital, United Kingdom. This regimen also achieved an impressive response rate of 71% in a phase II study involving 128 patients [6]. This led to comparison of ECF with FAMTX in a randomised phase III study involv- ing 274 patients with advanced oesophagogas- tric cancer. The response rate in this study with ECF was superior to FAMTX (45% versus 21%;

p = 0.0002) and overall survival was also supe-

rior (8.9 months versus 5.7 months; p = 0.0009).

This study also included patients with locally advanced inoperable disease as well as patients with metastatic disease. The response rate in patients with locally advanced disease was 58%

and a proportion of responding patients were able to proceed to surgery and subsequently underwent macroscopic and microscopic com- plete resection of the tumour (R0 resection).

A second randomised study has confirmed the activity of ECF. In this study ECF was com- pared with MCF, where MMC was combined with cisplatin and a larger dose of infused 5FU than is administered in ECF. In this study the response rates (42% versus 44%) and median survival time (9.4 months versus 8.7 months) for patients receiving both ECF and MCF were equivalent [7]. Whilst these measures of efficacy appeared equivalent, global quality of life was superior for patients receiving ECF. This is an

1111 2 3 4 5 6 7 8 9 1011 1 2 3 4 5 6 7 8 9 2011 1 2 3 4 5 6 7 8 9 3011 1 2 3 4 5 6 7 8 9 4011 1 2 3 4 5 6 7 8 9 5011 1 2 311

Table 26.2. Response rates achieved in phase II studies of

various chemotherapy agents in advanced oesophagogastric cancer

Chemotherapy Number of Response

agent patients rate (%)

5FU (infusion) 13 31

5FU (bolus) 392 21

Mitomycin-C 211 30

Epirubicin 61 26

Doxorubicin 68 25

Docetaxel 37 24

Paclitaxel 33 17

Irinotecan 60 23

Cisplatin 129 19

Carboplatin 57 5

Methotrexate 28 11

(3)

important measure for a treatment that is given with palliative intent and this has led to the con- clusion that ECF is the superior regimen.

ECF is the Most Active Combination Chemotherapy Regimen in

Oesophagogastric Cancer

The evidence therefore indicates that the ECF regimen is the most active combination chemotherapy regimen for patients with advanced oesophagogastric cancer. Although this regimen is widely accepted as standard treatment, its acceptance has not been univer- sal. The reluctance to accept ECF has arisen for a number of reasons and various criticisms of the ECF versus FAMTX study have been raised to account for this reluctance. Firstly, there was a perception that the patients receiving FAMTX in the trial comparing FAMTX with ECF had fared badly. Further randomised studies using FAMTX now suggest that the median survival achieved using FAMTX of approxi- mately 6 months is comparable to other studies.

Secondly, more patients receiving ECF pro- ceeded to a potentially curative surgical resec- tion, which may have biased the results of this study. Whilst many would regard this result likely to be due to the superior activity of ECF, the role of neoadjuvant ECF in patients with locally advanced disease has never been for- mally assessed in a randomised study. Thirdly, the administration of ECF requires the insertion of long-term indwelling venous catheters and results in a relatively modest prolongation of median survival in comparison with regimens such as FAMTX.

ECF achieves a modest median survival dura- tion in patients with advanced oesophagogastric cancer, with a median survival time of approx- imately 9 months. The relatively short survival times highlight the need for ongoing improve- ment in the treatment of patients with advanced oesophagogastric cancer. This is particularly the case for elderly and less fit patients for whom the side effects of intensive combination chemotherapy may be significant.

Future Studies

In order to attempt to improve the results achieved using chemotherapy, several ran-

domised studies incorporating novel agents are currently in progress. A multicentre study coor- dinated by the Royal Marsden Hospital is using ECF as the standard arm and is comparing this with substitution of cisplatin by the third gen- eration platinum agent oxaliplatin and substi- tution of 5FU with the oral fluoropyrimidine capecitabine. The use of capecitabine in this fashion avoids the requirement for indwelling venous catheters.

Novel agents with activity in oesophagogas- tric cancer include the taxanes and the topoiso- merase I inhibitor irinotecan (CPT11). Phase II studies have suggested that CPT11 plus 5FU is an active regimen (response rate 42%) [8].

Similarly, the combination of docetaxel, cis- platin and 5FU appears active (response rate 44%) [9]. These regimens are now being com- pared to standard treatment in randomised phase III clinical trials.

Biological Therapies

Further strategies currently being evaluated in an effort to improve the results in patients with advanced oesophagogastric cancer include the use of biological therapies directed at pathways perceived to be important in the pathogenesis of gastric cancer. One of these approaches involves the use of marimastat, a matrix metalloproteinase (MMP) inhibitor. The MMP enzymes constitute a family of proteolytic enzymes. Elevated levels of MMP enzymes have been associated with a poor prognosis in gastric cancer. A randomised study comparing mari- mastat with placebo in patients with gastric cancer demonstrated prolongation of failure- free survival and after protracted follow-up an overall survival advantage, especially for those patients who had received prior chemotherapy.

However, the prolongation in survival was modest at approximately 1 month and overall quality of life was not improved [10]. Therefore further evaluation of the role of marimastat is likely to be required in order to define a role for this drug in patients with advanced gastric cancer.

Other possible approaches include agents tar- geted to the epidermal growth factor receptor (EGFR). Antibodies to EGFR and drugs inhibit- ing the EGFR kinase domain are currently being evaluated in oesophagogastric cancer as well as other tumour types.

111

2

3

4

5

6

7

8

9

1011

1

2

3

4

5

6

7

8

9

2011

1

2

3

4

5

6

7

8

9

3011

1

2

3

4

5

6

7

8

9

4011

1

2

3

4

5

6

7

8

9

5011

1

2

311

(4)

Adjuvant Therapy

Although a proportion of patients may be cured by surgical resection of primary oesophageal or gastric tumours, 70% or more of patients will subsequently develop recurrent disease. This is due to the presence of unresected micrometas- tasis. Adjuvant treatment aims to eliminate residual tumour cells and increase the propor- tion of patients achieving long-term disease- free intervals and overall survival benefit.

Instituting treatment soon after surgery may have a number of advantages. The tumour burden is low and the likelihood of development of chemotherapy-resistant cell clones smaller.

The knowledge of the range of agents with activ- ity in advanced disease raises the possibility of utilisation of these agents in the adjuvant setting.

Gastric Cancer

The majority of studies of adjuvant therapy after resection of gastric cancers have failed to demonstrate any benefit for this treatment. A number of meta-analyses have also failed to indicate any benefit for adjuvant therapy, although a more recent analysis has suggested that there may be a small benefit [11]. An important caveat in interpretation of the data is that many of the studies used suboptimal adju- vant chemotherapy regimens, with many involving treatment using single chemotherapy agent regimens. In addition, many of the studies included relatively small numbers of patients and therefore there would need to be substan- tial differences in outcome for these to be sta- tistically significant. Adjuvant chemotherapy is therefore regarded as an unproven treatment after surgical resection of gastric cancer and eli- gible patients should be considered for adjuvant chemotherapy trials.

Oesophageal Cancer

Adjuvant chemotherapy after surgical resection of squamous cell carcinoma of the oesophagus has been evaluated in two randomised studies using the combinations of cisplatin with vindesine and cisplatin with 5FU given for two cycles [12–14]. In each study, there was no impact on overall survival for the group receiv- ing chemotherapy. Like the results of the gastric

cancer trials, relatively small numbers of patients have been studied and the chemo- therapy regimens are not necessarily optimal.

For example, it is unlikely that two cycles of chemotherapy would have a dramatic impact on micrometastatic disease. However, at this stage it must be concluded that postoperative adju- vant chemotherapy remains an unproven treat- ment for oesophageal carcinoma.

Adjuvant

Chemoradiotherapy

Gastric Cancer

Since both local as well as systemic recurrence represents a common problem after gastric cancer resection, a North American Intergroup trial evaluated a combined modality adjuvant regimen compared with observation. This incorporated postoperative chemotherapy involving one cycle of 5FU and leucovorin (LV) followed by two cycles of 5FU/LV administered concurrently with radiotherapy followed by two further cycles of 5FU/LV. The schema of treat- ment is outlined in Figure 26.1 [15]. The results of this adjuvant treatment showed that this therapy led to a 10% survival advantage after 3 years (52% versus 41%; p = 0.03). Failure-free survival was 49% for the treatment arm com- pared with 32% for no adjuvant therapy (p = 0.001). Whilst this treatment is now regarded as standard therapy in North America, it has not yet been widely accepted in Europe because of concerns about toxicity with abdom- inal chemoradiation and the adequacy of surgery. In the trial 41% and 32% of patients suffered grade 3 and 4 toxicities respectively and 54% of patients received less than a D1 resec- tion.

Neoadjuvant Treatment

Neoadjuvant therapy involves the administra- tion of systemic treatment before loco-regional therapy such as surgery. This approach to the treatment of oesophagogastric cancer offers a number of potential advantages. It allows systemic treatment to be delivered as early as possible, which may have advantages from the

1111

2

3

4

5

6

7

8

9

1011

1

2

3

4

5

6

7

8

9

2011

1

2

3

4

5

6

7

8

9

3011

1

2

3

4

5

6

7

8

9

4011

1

2

3

4

5

6

7

8

9

5011

1

2

311

(5)

perspective of treatment of micrometastatic disease. In addition, it allows the possibility that inoperable locally advanced tumours may be downstaged, thereby rendering these tumours amenable to potentially curative surgical resec- tion. It also provides prognostic information, with tumour response to neoadjuvant chemotherapy representing the most important prognostic factor for prolonged survival in gastric cancer [16]. The principal disadvantage of neoadjuvant chemotherapy is the possibility that tumour progression may occur during the course of treatment, thereby rendering the tumour inoperable and allowing the risk of dis- semination of disease.

Gastric Cancer

Phase II studies have demonstrated that neoad- juvant chemotherapy is an effective strategy in selected patients with gastric cancer. A current Medical Research Council (MRC) study is com- paring the results after three cycles of ECF neoadjuvant chemotherapy followed by surgical resection followed by a further three cycles of ECF adjuvant chemotherapy with surgical resection alone for patients with cancers of the stomach or oesophagogastric junction. The design of this trial is illustrated in Figure 26.2.

This study is close to completing accrual and should provide valuable information about the role of neoadjuvant chemotherapy in gastric cancer.

Oesophageal Cancer

In the case of oesophageal cancer, preopera- tive approaches have included neoadjuvant chemotherapy or neoadjuvant chemoradiation.

The neoadjuvant chemotherapy studies have often included relatively small numbers of patients receiving a variety of chemotherapy regimens. However, two large studies have recently been reported, one of which has shown a significant benefit for this approach. The results of all these studies are summarised in Table 26.3. A recent MRC study involving 800 randomised patients compared preoperative chemotherapy using two cycles of cisplatin 80 mg/m

2

and 5FU 1000 mg/m

2

d1–4 with no preoperative treatment [17]. The trial design is illustrated in Figure 26.3. Preoperative chemotherapy was feasible and resulted in a higher rate of macroscopic complete resection (84% versus 71%). The R0 resection rate was 60% in the chemotherapy arm compared with 55% in the surgery arm. This constitutes an important outcome, as only patients who achieve an R0 resection would be anticipated to be able to achieve long-term survival. Impor- tantly, there was no increase in perioperative complications or deaths after preoperative chemotherapy. There was a statistically signifi- cant improvement in both progression-free and overall survival, with 2-year survival being 45%

in the combined modality arm compared with 35% in the surgery alone arm.

111 2 3 4 5 6 7 8 9 1011 1 2 3 4 5 6 7 8 9 2011 1 2 3 4 5 6 7 8 9 3011 1 2 3 4 5 6 7 8 9 4011 1 2 3 4 5 6 7 8 9 5011 1 2 311

Figure 26.1. Outline of trial comparing postoperative chemoradiotherapy with observation after resection of gastric cancer.

(6)

A similar schedule of preoperative treatment has also been evaluated in a large American study [18]. This trial compared the results of surgery after preoperative chemotherapy using three cycles of cisplatin and fluorouracil with surgery alone. Preoperative chemotherapy achieved a small although not statistically sig- nificant improvement in the R0 resection rate.

However, this did not result in any impact on overall survival.

It is difficult to explain the different results from these two studies. Nevertheless, the large number of patients enrolled in the MRC study makes the results of this trial quite compelling.

It is likely that there are subgroups of patients who derive most benefit from such a neoadju- vant treatment approach. These may be identi- fied based on more accurate staging techniques such as endoscopic ultrasound or based on the molecular genotype of the tumour. However,

1111 2 3 4 5 6 7 8 9 1011 1 2 3 4 5 6 7 8 9 2011 1 2 3 4 5 6 7 8 9 3011 1 2 3 4 5 6 7 8 9 4011 1 2 3 4 5 6 7 8 9 5011 1 2 311

Table 26.3. Results of randomised studies of neoadjuvant chemotherapy in oesophageal carcinoma

Median overall survival (months)

Trial Number of Histology Chemotherapy Chemotherapy Surgery

patients

Roth et al 39 Squamous Cisplatin, vindesine, bleomycin 8 9

Nygaard et al 91 Squamous Cisplatin, bleomycin 8 8

Schlag et al 77 Squamous Cisplatin, 5FU 3 cycles 10 10

Kok et al 160 Squamous Cisplatin, etoposide 2–4 cycles 18.5* 11

Kelsen et al 440 Squamous/ Cisplatin, 5FU 3 cycles 14.9 16.1

adenocarcionma

MRC OEO2 802 Squamous/ Cisplatin, 5FU 17.6* 13.6

adenocarcinoma

*p< 0.01.

Figure 26.2. Outline of MRC study evaluating neoadjuvant chemotherapy in operable gastric cancer.

(7)

until these groups are identified it would be rea- sonable to consider neoadjuvant chemotherapy in patients who were being considered for radical resection of oesophageal carcinoma.

Neoadjuvant

Chemoradiotherapy

Oesophageal Cancer

A randomised study has compared chemora- diotherapy with radical radiotherapy as defini-

tive treatment for oesophageal carcinoma.

This study compared two cycles of cisplatin 75 mg/m

2

and 5FU 1000 mg/m

2

d1–4 given con- currently in weeks 1 and 5 with 50 Gy radio- therapy followed by two further cycles of the same chemotherapy with 64 Gy radiotherapy alone (Figure 26.4) [19]. The majority of cases were squamous cell carcinomas, but adenocar- cinomas were included. Median survival was 14 months in the combined modality arm compared with 9 months in the radiotherapy arm. The 5-year survival rate was 27% for the combined modality therapy versus 0% for radiotherapy.

111 2 3 4 5 6 7 8 9 1011 1 2 3 4 5 6 7 8 9 2011 1 2 3 4 5 6 7 8 9 3011 1 2 3 4 5 6 7 8 9 4011 1 2 3 4 5 6 7 8 9 5011 1 2 311

Figure 26.3. Outline of MRC study evaluating neoadjuvant chemotherapy in operable oesophageal cancer.

Figure 26.4. Outline of trial comparing radical radiotherapy with chemoradiotherapy as definitive treatment for oesophageal cancer.

(8)

The positive results of this study have led to acceptance of chemoradiotherapy as a possible treatment option for those patients who are not surgical candidates. This may apply for those patients who are not candidates for surgical resection on the grounds of concurrent medical disorders or because the anatomical location of the primary tumour renders surgery technically difficult, such as tumours arising in the cervical oesophagus. Nevertheless, the local failure rate with such an approach is significant (47%).

This has stimulated trials which incorporate chemoradiotherapy in addition to surgery.

Whilst a variety of regimens have been studied, most randomised studies evaluating these regimens have been negative. These results are summarised in Table 26.4. Whilst the study by Walsh et al has reported improved sur- vival with neoadjuvant chemoradiotherapy prior to surgery compared with surgery alone, this study has been criticised because of the relatively poor outcomes achieved in the surgery alone control arm [20]. A limitation of chemoradiotherapy is toxicity, which is increased with such therapy. For instance, in the trial reported by Urba et al, neutropenic sepsis occurred in 39% of patients and 63% of patients required feeding tubes in order to maintain nutrition because of radiation oesophagitis [21].

Therefore, in the absence of any significant benefit attributable to this approach, neoadju- vant chemoradiation prior to surgery for oesophageal carcinoma is currently regarded as an unproven treatment.

Improvements in Adjuvant and Neoadjuvant

Therapies

As new agents are evaluated further in the advanced setting, it is likely that those agents with the highest activity will be used in the adju- vant and neoadjuvant setting. However, even the use of these agents is unlikely to benefit all patients. It is likely that only selected patients will derive benefit from adjuvant or neoadju- vant therapy. These are patients who only have micrometastatic disease and whose tumours are responsive to chemotherapy.

Exclusion of Patients with Metastatic Disease

Conventional staging procedures do not exclude a number of patients with metastatic disease. However, the use of positron emission tomography (PET) scans has been shown to provide a useful adjunct to conventional staging using CT scans and endoscopic ultrasound examination [22]. The inclusion of PET scans could allow radical surgery and chemotherapy to be targeted at those patients who are most likely to benefit.

1111 2 3 4 5 6 7 8 9 1011 1 2 3 4 5 6 7 8 9 2011 1 2 3 4 5 6 7 8 9 3011 1 2 3 4 5 6 7 8 9 4011 1 2 3 4 5 6 7 8 9 5011 1 2 311

Table 26.4. Results of randomised studies of neoadjuvant chemoradiotherapy in oesophageal carcinoma

Trial Number Histology Chemotherapy Radiotherapy Chemotherapy Surgery

of patients dose

Le Prise et al 86 Squamous Cisplatin, 5FU 20 Gy, 10 10 10.5

fractions

Nygaard et al 88 Squamous Cisplatin, bleomycin 35 Gy in 20 9 8

fractions

Bosset et al 282 Squamous Cisplatin, 2 cycles 37 Gy in 10 18.6 fractions

Walsh et al 113 Adenocarcinoma Cisplatin, 5FU 2 cycles 40 Gy in 15 16*

fractions

Urba et al 100 Squamous/ Cisplatin, 45 Gy in 30 16.9

adenocarcinoma vinblastine, 5FU 2 cycles fractions

*p< 0.01.

(9)

Prediction of Tumour Response Using Molecular Markers

There has been considerable interest in the use of molecular markers to identify tumours which are most likely to respond to chemotherapy.

This has not been extensively evaluated in oesophagogastric cancers. However, studies involving small numbers of patients with gastric cancer showed that patients whose tumours had elevated levels of the enzyme thymidylate syn- thase (TS) were resistant to 5FU. Similarly, tumours with elevated levels of mRNA encod- ing the DNA repair gene ERCC1 were more likely to be resistant to cisplatin [23]. Further evaluation of the role of molecular markers may allow the identification of patients who are most likely to benefit from chemotherapy, and who could potentially be targeted to receive adjuvant or neoadjuvant chemotherapy. This type of strategy is advantageous as it ensures that only those patients who are likely to derive benefit from chemotherapy actually receive that treat- ment, thereby sparing toxicity in those who do not derive benefit.

Conclusions

Combination chemotherapy is proven to benefit patients with advanced oesophagogastric cancer. It is hoped that improvements in the treatment of advanced stage disease together with careful case selection will allow more patients with early stage disease to benefit from adjuvant or neoadjuvant chemotherapy or chemoradiotherapy.

Questions

1. What are the goals of palliative chemotherapy in advanced oesopha- gogastric cancer?

2. Which combination chemotherapy regimen is considered most active in advanced oesophagogastric cancer?

3. Which adjuvant therapy has shown a survival advantage after resection of gastric cancer?

4. What are the potential advantages and disadvantages of neoadjuvant chemo- therapy in oesophageal cancer?

5. Which neoadjuvant chemotherapy regi- men has achieved a survival advantage in operable oesophageal cancer?

References

1. Glimelius B, Hoffman K, Graf W et al. Cost-effectiveness of palliative chemotherapy in advanced gastrointestinal cancer. Ann Oncol 1995;6(3):267–74.

2. Hill ME, Cunningham D. Medical management of advanced gastric cancer. Cancer Treat Rev 1998;24(2):

113–18.

3. Douglass HO, Jr, Lavin PT, Goudsmit A et al. An Eastern Cooperative Oncology Group evaluation of combina- tions of methyl-CCNU, mitomycin C, Adriamycin, and 5-fluorouracil in advanced measurable gastric cancer (EST 2277). J Clin Oncol 1984; 2(12):1372–81.

4. Wils JA, Klein HO, Wagener DJ et al. Sequential high- dose methotrexate and fluorouracil combined with dox- orubicin – a step ahead in the treatment of advanced gastric cancer: a trial of the European Organization for Research and Treatment of Cancer Gastrointestinal Tract Cooperative Group. J Clin Oncol 1991; 9(5):

827–31.

5. Vanhoefer U, Rougier P, Wilke H et al. Final results of a randomized phase III trial of sequential high-dose methotrexate, fluorouracil, and doxorubicin versus etoposide, leucovorin, and fluorouracil versus infu- sional fluorouracil and cisplatin in advanced gastric cancer: A trial of the European Organization for Research and Treatment of Cancer Gastrointestinal Tract Cancer Cooperative Group. J Clin Oncol 2000;18(14):2648–57.

6. Findlay M, Cunningham D, Norman A et al. A phase II study in advanced gastro-esophageal cancer using epirubicin and cisplatin in combination with continu- ous infusion 5-fluorouracil (ECF). Ann Oncol 1994;

5(7):609–16.

7. Ross P, Nicolson M, Cunningham D et al. A prospective randomised trial comparing MCF with ECF in advanced oesophago-gastric cancer. Submitted 2001.

8. Pozzo C, Bugat R, Peschel C et al. Irinotecan in combi- nation with CDDP or 5FU and folinic acid is active in patients with advanced gastric or gastro-oesophageal junction adenocarcinoma: Final results of a ran- domised phase II study. Proc Am Soc Clin Oncol 2001;

20(abstr).

9. Van Cutsem E, Ayani J, Tjulandin S et al. Docetaxel in combination with cisplatinum with or without 5-fluo- rouracil in patients with advanced gastric or GE junc- tion adenocarcinoma: Preliminary results. Ann Oncol 2001;11(S4)(abstr).

10. Fielding J, Scholefield J, Stuart R et al. A randomized double-blind placebo-controlled study of marimastat in patients with inoperable gastric adenocarcinoma. Proc Am Soc Clin Oncol 2000;19(abstr).

111

2

3

4

5

6

7

8

9

1011

1

2

3

4

5

6

7

8

9

2011

1

2

3

4

5

6

7

8

9

3011

1

2

3

4

5

6

7

8

9

4011

1

2

3

4

5

6

7

8

9

5011

1

2

311

(10)

11. Mari E, Floriani I, Tinazzi A et al. Efficacy of adjuvant chemotherapy after curative resection for gastric can- cer: a meta-analysis of published randomised trials.

A study of the GISCAD (Gruppo Italiano per lo Studio dei Carcinomi dell’Apparato Digerente). Ann Oncol 2000;11(7):837–43.

12. Webb A, Cunningham D, Scarffe JH et al. Randomized trial comparing epirubicin, cisplatin, and fluorouracil versus fluorouracil, doxorubicin, and methotrexate in advanced esophagogastric cancer. J Clin Oncol 1997;15(1):261–7.

13. Ando N, Iizuka T, Kakegawa T et al. A randomized trial of surgery with and without chemotherapy for localized squamous carcinoma of the thoracic esophagus: the Japan Clinical Oncology Group Study. J Thorac Cardiovasc Surg 1997;114(2):205–9.

14. Ando N, Iizuka T, Kakegawa T et al. A randomized trial comparing surgery to surgery plus postoperative chemotherapy for localised squamous cell carcinoma of the thoracic esophagus. Proc Am Soc Clin Oncol 1998;17. (abstr).

15. Macdonald JS, Smalley S, Benedetti J et al. Postoperative combined radiation and chemotherapy improves dis- ease-free survival (DFS) and overall survival (OS) in resected adenocarcinoma of the stomach and GE junc- tion. Results of Intergroup Study INT-0116 (SWOG 9008). Proc Am Soc Clin Oncol 2000; 19 (abstr).

16. Lowy AM, Mansfield PF, Leach SD et al. Response to neoadjuvant chemotherapy best predicts survival after curative resection of gastric cancer. Ann Surg 1999;229(3):303–8.

17. Clark P. Surgical resection with or without pre-opera- tive chemotherapy in oesophageal cancer: an updated analysis of a randomised controlled trial conducted by the UK Medical Research Council Upper GI tract cancer group. Proc Am Soc Clin Oncol 2001;20(abstr).

18. Kelsen DP, Ginsberg R, Pajak TF et al. Chemotherapy followed by surgery compared with surgery alone for localized esophageal cancer. N Engl J Med 1998;339(27):

1979–84.

19. Herskovic A, Martz K, al Sarraf M et al. Combined chemotherapy and radiotherapy compared with radio- therapy alone in patients with cancer of the esophagus.

N Engl J Med 1992;326(24):1593–8.

20. Walsh TN, Noonan N, Hollywood D et al. A comparison of multimodal therapy and surgery for esophageal ade- nocarcinoma. N Engl J Med 1996;335(7):462–7.

21. Urba SG, Orringer MB, Turrisi A et al. Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 2001;19(2):305–13.

22. Flamen P, Lerut A, Van Cutsem E et al. Utility of positron emission tomography for the staging of patients with potentially operable esophageal carci- noma. J Clin Oncol 2000;18(18):3202–10.

23. Metzger R, Leichman CG, Danenberg KD et al. ERCC1 mRNA levels complement thymidylate synthase mRNA levels in predicting response and survival for gastric cancer patients receiving combination cisplatin and fluorouracil chemotherapy. J Clin Oncol 1998;16(1):

309–16.

1111

2

3

4

5

6

7

8

9

1011

1

2

3

4

5

6

7

8

9

2011

1

2

3

4

5

6

7

8

9

3011

1

2

3

4

5

6

7

8

9

4011

1

2

3

4

5

6

7

8

9

5011

1

2

311

Riferimenti

Documenti correlati

system of adiponectin (ADN), an insulin-sensitizing and anti- inflammatory adipokine, in tissues known to be the target of chronic diseases associated to obesity

TFF1 is mainly associated with breast cancer and gastric cancer (GC), but have been described changes in expression levels also in pancreatic cancer, lung, prostate and

Endoscopy won’t localize lesion Pt will have had prior ulcer surgery Pt will have had prior AAA repair All coags will be abnormal NGT won’t get bilious return. Bleeding will be

2.3.3 Daphnia magna: acute and reproduction toxicity tests The 48 h daphnid toxicity test was conducted according to OECD test guideline 202.. magna Straus clone neonates between 6

In this paper we propose an output-feedback Model Predictive Control (MPC) algorithm for linear discrete-time systems affected by a possibly unbounded additive noise and subject

Phase II study of first-line LY231514 (multi-targeted antifolate) in patients with locally advanced or metastatic colo- rectal cancer: An NCIC Clinical Trials Group study. John W,

Importantly, not only ground states but also the low-energy part of the spectrum of systems described by CFT shows sub-volume (logarithmic) scaling of the entanglement entropy.. We

The
 aim
 of
 this
 work
 is
 to
 identify,
 to
 describe
 and
 to
 explain,
 by
 analytical
 and
 stratigraphic
 studies
 (that
 implied
 field