Skip to main content

Main menu

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart
  • Log out

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Subscriptions

Chemotherapy in small cell lung cancer

K. Østerlind
European Respiratory Journal 2001 18: 1026-1043; DOI: 10.1183/09031936.01.00266101
K. Østerlind
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Chemotherapy is the backbone in the treatment of small cell lung cancer (SCLC) and radiotherapy is an important adjunct in limited stage disease. The role of chest irradiation is now documented in three meta-analysis, based on the same body of data. Trials on timing, scheduling and fractionation could have followed a more stringent development line but altogether, the highest efficacy seems to be obtained with early, concurrent twice-daily chest irradiation. Patients in complete remission should have prophylactic cranial irradiation, which reduces the risk of brain metastases and of death from SCLC.

Four series of chemotherapy seem to be sufficient in limited-stage disease while six is recommended in extensive disease. The combination of etoposide plus cis- or carboplatin is appropriate in both stages and addition of other agents has no clinically important impact on the survival. Use of haematological growth factors such as granulocyte colony stimulating factor (G-CSF) and granulocyte macrophage colony stimulating factor (GM-CSF) may enable higher doses or more frequent dosage. Three randomized trials on GM-CSF showed a negative outcome while G-CSF support may result in better survival rates, but a more cost-efficient policy must be found. High-dose chemotherapy plus haematological stem-cell support is still under investigation but disappointing long-term survival rates means there is not much optimism for this strategy.

New strategies in general are requested in the treatment of extensive-stage disease and of elderly patients. Phase II trials suggest that good-risk patients with extensive disease should be treated aggressively, intermediate-risk patients more gently, and palliation must be the primary aim in the treatment of poor-risk patients. In elderly patients impressive survival rates are obtained with 3–4 series of chemotherapy and radiation delivered in 5–10 fractions.

A number of new agents are active but more trials are required before each has found a place, if any, in the treatment of small cell lung cancer. To conclude, the randomized trial is still an important instrument in clinical oncology, and trials in small cell lung cancer must be large, which is why the cooperation of organizations and multicentres is urgent.

  • chemotherapy
  • elderly
  • growth factor
  • radiotherapy
  • randomized trial
  • small cell lung cancer

Chemotherapy is the keystone in the treatment of small cell lung cancer (SCLC). Objective remission and good palliation is achieved in ∼80% of the patients, but the remissions are in general short (mean <1 yr), and few are cured. As an example, 5- and 10-yr survival rates, in 1,714 SCLC patients treated in trials in Copenhagen during 1973–1987, were only 3.5 and 1.8%, respectively 1. Only 184 (11%) of these patients received chest irradiation. In those days the role of radiotherapy was still an issue of debate, but two meta-analyses, published in 1992 2, ended the discussion. Search for an optimal schedule and timing of the radiotherapy has prompted several randomized trials and the sum of the data from these trials suggests that radiotherapy should be given early, concurrently and twice daily 3. Irradiation of the adrenals has never proved worthwhile 4 while prophylactic cranial irradiation (PCI) in complete remission patients has a significant positive impact on the prognosis, proven in a meta-analysis 5. Trials aiming to find the optimal dose and indications for PCI are under way.

Sixty to 65% of patients with SCLC have extensive disease at the time of diagnosis. Spread of disease prevents appropriate limits for a chest radiation field, and the prognosis is significantly reduced. Many efforts have been undertaken to improve treatment outcome in extensive stage disease, especially trials on high dose or dose-accelerated regimes, but the outcome has been disappointing. High-dose chemotherapy plus haematological stem-cell support has found a place in haematology, such as in the treatment of recurrent lymphomas, and the strategy has also been investigated as a first-line treatment for good-prognosis patients with SCLC, mostly in phase II trials. High complete response rates are obtained, but there is considerable toxicity and the long-term survival rates are not improved.

Twenty-five per cent of patients with SCLC are aged >74 yrs and most of these patients may not tolerate combination chemotherapy. But elderly people are less fatalistic today, they expect treatment for conditions such as cancer, and although there have been some trials on older patients during the last 10–15 yrs, the treatment options are limited.

The fact that age has not proved to be an important prognostic factor in a large series 6 suggests that treatment for cure may be a reasonable policy in some elderly patients. The prognosis in SCLC is influenced by stage of disease plus other clinical characteristics such as performance status (PS) and plasma lactate dehydrogenase (LDH), followed by plasma sodium, anaemia, plasma albumin, alkaline phosphatase, and bicarbonate 6–9. Prognostic factors, especially PS and LDH, are important as in- and exclusion criteria in treatment trials, as stratification or balancing variables in randomized trials, and as potential end point confounders. In that context further standardization is warranted, a task which has recently been undertaken by a staging committee under the International Association for the Study of Lung Cancer (IASLC).

Surgery is seldom possible in SCLC 10 and explorative thoracotomies are rare today due to routine use of percutaneous lung biopsies and modern imaging techniques to guide invasive procedures if necessary. In addition mediastinoscopy has had a renaissance in the staging of lung cancer 11. More than 85% of SCLC patients have mediastinal lymph-node metastases 12 and in a time when better local control of the tumour seems possible, due to improved radiotherapy most will agree that surgery has had its role in the treatment of SCLC.

A number of new cytotoxic drugs have been investigated in the last decade including an antimetabolite (gemcitabine), taxanes and topoisomerase I inhibitors. Although the trials have resulted in reasonable developments, a complete understanding of the best way to use these agents is needed.

Treatment strategies in small cell lung cancer

Clinical research in to the treatment of SCLC is not driven by the same optimism as it was 20-yrs-ago but trials on new agents and new biological principles in conjunction with the classical regimes are always under way. With trials carried out by laboratories, who never lose interest, or initiated by drug companies, often in an international set up. Local trials on feasibility and practical, clinical characteristics of treatment regimes for SCLC also occur, often in certain categories such as elderly or poor-risk patients. Survival is still the major end point in trials on SCLC but outcome variables also include measures of palliation, reduced toxicity, days of hospitalization and cost-efficacy ratio. New strategies, based on a fast developing, molecular biological understanding of cancer, will certainly get an increasing impact on the clinical drive for a better treatment. Biological strategies have been tested in clinical practice since the introduction of chemotherapy three-decades ago. The classical example is the Goldie-Coldman hypothesis 13 from which a computer growth simulation model showed that alternating chemotherapy with noncrossresistant regimes would increase the chances of a successful outcome. The principle, however, turned out to have only a restricted impact in SCLC, detectable in extensive but not in limited stage disease 14–16.

Intensive and high-dose chemotherapy has found a place in the treatment of haematological neoplasms and achievements from this field plus development of the blood stem-cell rescue technology have stimulated trials on this strategy in solid tumours, including SCLC. Whether or not high-dose chemotherapy has a role in the treatment of SCLC is still an open question.

Radiotherapy is now a standard in the treatment of limited stage SCLC. Current investigations focus on the timing, fractionation and interaction with concurrent chemotherapy. PCI reduces the risk of brain metastases 17. The efficacy seems to be dose dependant and the balance point between maximal effect and minimal-late toxicity has to be found.

New cytotoxic drugs are steadily being investigated in various combination regimens, often in trials organized by the manufacturing drug company. The steering of this process is a big challenge to all clinical oncologists. Similar conditions are typical for clinical investigations on biological processes activated by the cancer to enable invasion, nourishment and spread, which include proteolysis, angiogenesis and interaction with the immune system.

Treatment of limited disease

Evidence for the usefulness of chest irradiation in limited-stage SCLC was established in two meta-analyses published in 1992 2, 18. The body of data for the two investigations was essentially the same, i.e. outcome of randomized trials from the period 1975–1989 including a total of some 2000 patients. One group retrieved the raw data 2 while the other skimmed the publications. The outcomes, however, were much alike, both proving a significant improvement in survival. As an example, the 3-yr survival rates in Pignon's study were 14.3% in 1,111 patients assigned to combined modality therapy compared to 8.9% in 992 patients treated with chemotherapy alone. The 3-yr survival rates in recent trials are ∼20% and rates >30% are reported. Several factors may contribute to higher rates. 1) Stage migration as a result of new imaging technologies in staging of lung cancer such as spiral computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography (PET) scanners. 2) Chemotherapy in recent trials is based on the combination of cisplatin+etoposide (PE), which is more efficacious than some of the odd combinations used in the 1970–1980s 19 and less toxic than regimes including doxorubicine when delivered concurrently with the irradiation. 3) Most trials today include prophylactic brain irradiation to complete responders. 4) Timing, dosage, fractionation and computer-assisted CT based simulation of the chest irradiation may each contribute to the better long-term survival rates seen today.

Timing, dosage and fractionation of the chest irradiation are not new issues in SCLC. Early trials, however, were in general unable to give clear answers. The advantages anticipated were too optimistic so the trials were underdimensioned. Even the meta-analysis 2 was unable to prove significant differences in survival related to early versus late radiotherapy or to sequential versus nonsequential regimes.

Establishment of nationwide or international joint trials is the key to the recruitment of enough patients. The Canadian Lung Cancer Group is an example to follow and one of the questions investigated by the Canadian group is the role of timing of the chest irradiation in relation to the course of chemotherapy of limited-stage SCLC 20. A total of 308 patients were randomized to early (week 3) or late (week 15) irradiation concurrent with PE. The two agents were alternated with cyclophosphamide, doxorubicine and vincristine (CAV) for a total of six cycles of chemotherapy. The dose of radiotherapy was 40 Gy in 15 fractions over 3 weeks. Both progression-free and overall survival were significantly superior in the early radiotherapy arm. Thus, overall survival after 2 yrs was 40% versus 34% (tables 1⇓ and 2⇓), and after 3 yrs 30% versus 22%. The cumulative proportions of local recurrence did not differ, however, exceeding 50% in both arms. In a Danish trial 21 199 patients were randomized to early (weeks 1, 2, 4 and 5) or late (weeks 19, 20, 24, 25) radiotherapy. A course of PE was given in week 3 and week 22, respectively and altogether the chemotherapy comprised 3 cycles PE+6 cycles CAV. The first cycle of chemotherapy was delayed for 2 weeks in the “early” arm. There was no difference in survival between the arms. The 2-yr survival rates were 20 and 19%, respectively.

View this table:
  • View inline
  • View popup
Table 1—

OlgpØyHKtCvØCedtCptrlcdc rlUdte.Ørlin rØ,dsØo

View this table:
  • View inline
  • View popup
Table 2—

OlgpØy HktCvØCedtCeptrlcdc rlUdte.Ørlin rØ,dsØoactCedC: ØU-

The Danish trial delayed chemotherapy in the “early” arm to avoid concurrent treatment. PE can, however, safely be given concurrently with radiotherapy, in contrast to the anthracyclines, which significantly increases the normal tissue radiotoxicity. This problem was clearly revealed in a North American multicentre trial 24 randomizing 386 patients to receive six-series CAV, given on day 1, every 3 weeks with or without concurrent chest irradiation: 30 Gy in 10 fractions weeks 1 and 2, plus 15 Gy in five fractions in week 7. Irradiation plus CAV increased the treatment toxicity, especially the haematological: 60% of the patients had grade 4 neutropenia compared to 39% in the other arm. There were six treatment-related deaths compared to three in the chemotherapy-alone arm. Only 57% of the patients received at least 80% of the intended dosage of the two myelotoxic agents (C and A) compared to 79% of the patients in the chemotherapy-alone arm. There was no significant difference in overall survival between the two arms but 2-yr survival in irradiated patients was 33% compared to 24% in nonirradiated patients.

The question of early versus late and of alternating versus sequential chest irradiation was addressed in a European multicentre trial (EORTC) randomizing 335 patients to early alternating or late sequential thoracic irradiation. The chemotherapy: five courses cyclophosphamide, doxorubicine, and etoposide (CAE) was not given concurrently. In the early arm, 50 Gy was given in weeks 6, 10, 14 and 18 alternating with CAE while patients in the late arm received 50 Gy at weeks 14–17, i.e. sequentially given between two courses of CAE. The dosage of chemotherapy was the same in the two arms, and >95% of the scheduled doses could be given in both, but with a cyclus time of 3 weeks in the sequential arm, and 4 weeks in the alternating arm, the dose rate (mg·m−2·week−1) differed with as much as 31%. The relative risk of death was in favour of patients treated with the sequential regime (0.88) but the difference was not statistically significant. Grade 4 leukopenia occurred in 72% of patients in the alternating arm compared to 42% in the sequential arm.

Hyperfractionation may increase the efficacy of radiotherapy in SCLC. The theory is based on in vitro irradiation of SCLC cells, which do not exhibit the characteristic shoulder phenomenon reflecting repair of sublethal damage. Twice-daily radiotherapy (TDR) in weeks 2, 5 and 8 alternating with chemotherapy (PE) was investigated in an early pilot study 23 but the outcome did not differ remarkably from usual-treatment outcome at that centre neither in efficacy or toxicity. But a much larger, randomized intergroup trial 3 resulted in significantly higher survival rates in the TDR arm compared to the once-daily radiotherapy (ODR) arm. Chemotherapy: four series PE was given concurrently in both arms. The total dose of radiotherapy was 45 Gy in both regimes (tables 3 and 4⇓⇓). Kaplan-Meier curves of overall survival were superimposable the first year but then began to deviate, resulting in differences of 47% versus 41% after 2 yrs and 26% versus 16% after 5 yrs. The toxicity was similar in the two arms: grade 4 granulocytopenia in 60% and 59% of the patients, respectively, and grade 4 thrombocytopenia in 8% in both arms. Grade 3 oesophagitis was seen in 27 and 11%, respectively, reflecting that 45 Gy in TDR versus ODR are not biologically equivalent regimes. For comparison, in a trial 24 of CAV plus concurrent irradiation, 30 Gy in 10 fractions, the haematological toxicity was similar and grade 3 oesophagitis occurred in 13%, but rates of long-term survival were considerably lower.

View this table:
  • View inline
  • View popup
Table 3—

OlgpØyHOrdlpøC e7dcØ3Uldeptrlcdc drrlUdledtC

View this table:
  • View inline
  • View popup
Table 4—

OlgpØyHOrdlpøC e7dcØ3Uldeptrlcdc drrlUdledtCactCedØU

The oesophageal mucosa is early reacting normal tissue and acute oesophagitis (≥grade 4) is a major clinical problem when chemo- and radiotherapy are given concurrently. The dose-limiting toxicity of TDR concurrent with cisplatin, cyclophosphamide and etoposide in conventional doses was investigated by Choi et al. 29 in a phase I study. The maximum-tolerated dose (MTD) of TDR was 45 Gy in 30 fractions over 3 weeks compared to at least 70 Gy in 35 fractions over 7 weeks for daily radiotherapy. Doses above this MTD seem to be applicable, however, if chemotherapy doses are reduced.

Other policies of TDR: early versus late and early concurrent versus late sequential, were investigated in two randomized trials, a Yugoslavian-Japanese 25 and a Japanese 28, respectively. In the first trial 54 Gy TDR in 36 fractions was given early: week 1–4; or late: week 6–9, concurrently with carboplatin (30 mg·day−1) plus etoposide (30 mg·day−1). Grade 3–4 acute oesophagitis was observed in 29% of 52 patients and in 25% of 51 patients, respectively. Subsequently, the patients received four-series PE in conventional doses. Median survival was >2 yrs in both arms, and the 5-yr survival rates were 30 and 15%, respectively, favouring the early radiotherapy regime. The other trial 28 included 231 patients between 1991–1995. Patients in both arms received TDR 45 Gy in 3 weeks, initiated on day 2, concurrent with PE in arm C and after four-series PE in the sequential arm S. A total of four-series PE was given, every 4 weeks in arm C but three-weekly in arm S. Dosage was the same. Median survival was 27 and 20 months, respectively, and 5-yr survival rates were 24 and 18% (p=0.057). Myelotoxicity was more severe in arm C but the nonhaematological toxicity, including oesophagitis, was not significantly different between the two arms. In contrast to these promising data experience in a French trial 27 on TDR was more sinister. The 27 patients received 45 Gy in 30 fractions concurrently with the second of six series PE (tables 3 and 4⇑⇑). The 2-yr survival rate was only 25% and the toxicity was problematic. Thus, 18 patients (67%) had grade 3–4 neutropenia, and two died whilst febrile and neutropenic and grade 3–4 oesophagitis was observed in 50% of the patients.

In a North-American randomized trial 26 TDR was given in split-course: 16 fractions on days 1–10 plus 16 fractions on day 29–38 to a total of 48 Gy was not superior to once-daily irradiation of 50.4 Gy in 28 fractions over 6 weeks. The 2- and 3-yr survival rates were 47 and 34% for patients in the ODR arm and 45 and 29% for patients in the TDR arm (p=0.44). The toxicity level was acceptable and similar in the two arms except that there were significantly more patients with oesophagitis in the twice-daily arm: 12 versus 5%.

It may be concluded that TDR plus concurrent chemotherapy seems to be the best treatment for limited-stage SCLC leading to 5-yr survival in one-quarter of the patients. The regimen is, however, quite toxic for some patients. Optional patients are patients in the good-risk group characterized by a median survival of 20 months and a 2-yr survival rate of 40%. Staging procedures and other inclusion criteria applied in North American trials seem more appropriate for selection of this patient category than those used in European trials. And selection is important in order to reduce overtreatment and toxic complications in patients who, it must be anticipated, basically have poor chances of long-term survival. Still, the strategy of early radiotherapy in a twice-daily fractionation, concurrent with PE, needs further documentation in confirmatory trials. At the same time clinical investigations are needed to find a good treatment for limited-stage patients in whom the concurrent chemo- plus TDR is too toxic. Seen in a broader clinical perspective that is also a challenge.

Prophylactic cranial irradiation

Brain metastases presenting at time of diagnosis can be brought into remission with systemic chemotherapy 30 but systemic chemotherapy cannot prevent occurrence of brain metastases. About 20–30% of the patients develop brain metastases during therapy 31. Patients in complete remission at conclusion of chemotherapy, proved with restaging procedures including a brain CT scan, have a cumulative risk of 50–60% of developing brain metastases over the subsequent 2 yrs. Survival after occurrence of brain metastases is short: median 3–6 months, although long-term survival can be seen in patients, who remain in systemic remission 32. The rate of brain metastases occurring in complete responders after conclusion of chemotherapy can be reduced significantly if cranial irradiation: 24–36 Gy is given prophylacticly 17, 22, 33 but an observed positive impact on overall survival was not statistically significant in these trials. This problem was solved with a meta-analysis published in 1999 5. Survival and relapse data on 987 patients from seven randomized trials were analysed. A total of 526 patients had PCI while 461 were controls. All were in complete remission at time of randomization. A minority of 140 patients (14%) had extensive disease when the systemic chemotherapy was initiated. The cumulative incidence of brain metastases 3-yrs after randomization was 33% in the PCI group compared to 59% in the control group. The risk of death was reduced 16% (p<0.01) in the PCI group corresponding to an increase in the 3-yr survival rate from 15.3 to 20.7%. The survival benefit appeared after 1 yr and the difference between the Kaplan-Meier curves persisted beyond 6 yrs. An editorial in the same issue of the New England Journal of Medicine concluded that “it is now resonable to include prophylactic irradiation as part of the treatment in patients with limited small-cell lung cancer … and of patients with extensive disease who have isolated metastases and are in complete remission” 34. The patient's age had no influence on the outcome. An irradiation dose of 30–40 Gy led to a significant lower risk of brain metastases compared to 24–25 Gy but the difference in overall survival (death hazard 0.81 versus 0.88) was not significant. A European multicentre randomized trial has now been initiated to investigate if 30–40 Gy is better than the lower dose. With a statistical power of 75% and α=0.05 a total of 374 patients must be included in each arm. The outcomes of the meta-analysis have been reproduced in two new meta-analyses 35, 36. Meert et al. 35 were concerned about the paucity of data on possible late neurological impairment following PCI. Currently, the worried patient can be reassured that late toxicity after PCI, such as impairment of cognitive function, is less frequent than suggested after observations made with an early generation CT scanner 37. Three investigations 17, 22, 38 found few additional patients with abnormal neurological or neuropsychological findings as well as CT-scan changes 2 yrs after PCI compared to baseline examinations performed after chemotherapy and before randomization to PCI. At baseline, however, impairments of cognitive functions could be detected in up to 50% of the patients while only a few had CT abnormalities, e.g. 17% of 183 patients in the French study 17. New CT abnormalities occurred in 10% of the patients after PCI versus 3% in the control group (p=0.6). For the individual patient, of course, loss of function may be a great problem, and possible risk factors should therefore be clarified, such as age and previous vascular incidents. PCI should not be given concurrently with the systemic chemotherapy.

High-dose chemotherapy

Two decades after the first trials were initiated 39, 40 high-dose chemotherapy with stem-cell support is still an experimental treatment strategy in SCLC. It has not been possible to prove a clear dose-effect relationship in chemotherapy of SCLC 41, the number of patients included in high-dose trials are restricted, because the age and general physical condition of the patients with SCLC will often make high-dose chemotherapy with stem-cell support a risky policy. No large randomized trials have been performed and the data available are not especially promising 42–46 (table 5⇓). Patients with extensive-stage disease do not seem able to obtain longer relapse-free survival than obtained on conventional treatment regimes and rates of long-term survival in limited-stage patients do not differ remarkably from the rates obtained with four-series of PE plus twice-daily chest irradiation 3.

View this table:
  • View inline
  • View popup
Table 5—

Trials on high dose chemothaerapy plus haematological stem cell support

Granulocyte colony stimulating factor or granulocyte macrophage colony stimulating factor and dose intensity

G-CSF has well defined roles in high-dose chemotherapy where it is used for mobilization of peripheral blood stem cells (PBSC) and for stimulation of granulocyte regrowth after retransfusion of PBSC. It's potential role as a way of increasing dosage of chemotherapy and thereby efficacy without use of PBSC support has been investigated in a number of trials. Two almost identical randomized trials 47, 48 included a total of 341 SCLC patients (27–39% with limited-stage disease) who received a relatively aggressive combination of cyclophosphamide 1 g·m−1 , doxorubicine 50 mg·m−1 , and etoposide 120 mg·m−1 on days 1–3 plus or minus subcutaneous treatment with G-CSF on days 8–13. Neutropenia with fever occurred in 28 and 57% in the arms of a trial by Crawford et al. 47 and in 26 and 53% in a trial by Trillet-Lenoir 48, and the number of hospitalization days were significantly reduced in both studies. Response and survival rates, however, were not statistically different. The question of cost was not considered in these papers but Nichols et al. 49 made an effort to calculate the cost consequences of a clinical strategy including G-CSF. They reviewed a sample of unselected patients treated with standard-dose chemotherapy in the period 1987–1992. The incidence of neutropenic fever was 12% in the first cycle and 18% overall. Assumptions of the effectiveness of G-CSF were based on data from the Neupogen licensing trial. A policy of 25% dose reduction alone after episodes of neutropenic fever was cheapest, addition of G-CSF in chemotherapy cycles after an episode increased cost moderately with a factor of 1.4 while pre-emptive use of G-CSF with all courses of chemotherapy would increase cost with a factor 6.7 to a total of 1.29 million dollars·patient−1. A cost-effective and ethically appropriate policy could make use of a less-aggressive regime to patients with a risk of neutropenic fever in the first cycle of >25%, estimated from simple characteristics as age, LDH, stage of disease and performance status 50, 51.

In addition to G-CSF, GM-CSF can reduce white blood cell (WBC) nadir after chemotherapy. The feasibility and possible advantage of GM-CSF after concurrent chemo- and radiotherapy in limited stage SCLC was investigated in a randomized trial by the Southwest Oncology Group 52 (table 6⇓). Treatment was a combination of cisplatin 25 mg·m−1 on days 1–3 plus etoposide 60 mg·m−1 on days 1–3, and chest irradiation: 45 Gy during week 1–5. There were significantly more patients with life-threatening thrombocytopenia in the GM-CSF arm, more days in hospital, more who needed intravenous antibiotics, more with nonhaematological toxicity, and more toxic deaths. There was no significant difference in survival and both 2-yr and 4-yr survival rates were inferior in the GM-CSF arm as compared to the control arm. It was concluded that concurrent use of haematological growth factors plus chemotherapy and daily radiotherapy should be avoided.

View this table:
  • View inline
  • View popup
Table 6—

Impact of granulocyte macrophage colony stimulating factor and dose intensity in treatment of small cell lung cancer

The feasibility and efficacy of dose-intensification plus GM-CSF in extensive SCLC was investigated in a joint French trial 53 (table 6⇑). Same cumulative doses of etoposide, cyclophosphamide, epirubicine and cisplatin were given in four cycles in the GM-CSF arm and in six cycles in the control arm. Cyclus time was 28 days and GM-CSF was given on days 10–14. The actual median cumulative doses received were: 84% in the control arm versus 77% in the high-dose arm. There were five and eight toxicity-related deaths in the control arm and the GM-CSF arm, respectively. Grade 4 neutro- and thrombocytopenia within the first two cycles as well as survival data are summarized in table 6⇑. The trial was stopped at the first planned interims analysis when it was realized that GM-CSF did not enable dosage acceleration. It was concluded that the higher dose-intensity achievable with this regimen in the treatment of extensive stage SCLC had no positive impact on the treatment outcome.

Similarly, outcome was also negative in a joint European trial on increased dose-intensity plus GM-CSF in limited-stage SCLC 54. In a two-by-two factorial design patients were firstly randomized to vincristine, iphosphamide, carbaplatin, etoposide (VICE) chemotherapy every 3 weeks or every 4 weeks and secondly randomized to treatment with GM-CSF or not for 14 days in each cycle (table 6⇑). Radiotherapy was first delivered after completion of the planned six-cycles of chemotherapy. GM-CSF did not reduce the incidence of haematological complications and it had no significant impact on either response rates or survival. Chemotherapy every 3 weeks prolonged survival in both limited and extensive-stage disease, and this influence was significant in a Cox analysis also including stage and tumour size. GM-CSF, however, had no influence on the ability to maintain the higher-dose intensity.

G-CSF has been investigated in similar randomized trials investigating accelerated chemotherapy plus or minus G-CSF (table 7⇓). Woll et al. 55 used the VICE regimen and a policy where next cycle was delivered as soon as the WBC count was ≥3.0, i.e. there was no fixed-dose interval. Dose-intensity was high in both groups and significantly higher in the G-CSF arm, leading to more treatment-related deaths: six versus one. The 2-yr survival rate was best in the G-CSF arm: 32% versus 15%, but the survival difference was not statistically significant. The same outcome of a similar strategy was observed in a Japanese randomized trial on 227 extensive disease patients 56. Patients in the control arm received CAV alternating with EP in conventional dosage versus weekly cisplatin, oncovine, doxorubisine, etoposide (CODE) plus G-CSF in the investigative arm G (table 6⇑) (C: cisplatin 25 mg·m−2 weekly, O: vincristine 1 mg·m−2 in weeks 1, 2, 4, 6 DE: doxorubicin 40 mg·m−2+etoposide 80 mg·m−2 for 3 days in weeks 1, 3, 5, 7, 9). Dose-intensity in arm G was twice that in the control arm, four versus 0 toxic deaths were observed, the response rates were 84 versus 77% but no significant difference in survival was achieved: median survival being 11.6 months and 10.9 months. A slightly different strategy was applied in a large British trial (Thatcher et al. 57). Patients were randomized to receive (ACE) adriamycin 40 mg·m−2, cyclophosphamide 1,000 mg·m−2 and etoposide 100 mg·m−2 i.v. on days 1 and 240 mg·m−2 p.o. on days 2 and 3 every 3 weeks (control arm) or ACE every 2 weeks plus G-CSF subcutaneous for 14 days after each treatment. Patients in the accelerated arm survived longer, corresponding to a reduced hazard ratio of 0.80. The hazard reduction was the same in both limited- and extensive-stage disease. The Kaplan-Meier curves deviated beyond 12 months with 2 yr survival rates of 13 and 8%, respectively. Grade 3–4 neutropenia was much less frequent in the G-CSF arm (table 7⇓) but more blood transfusions were given. The use of G-CSF added significantly to the cost of the therapy and it is necessary to find a more cost-effective way of using G-CSF. The trial showed, that dose intensification is possible, and that it seems to have a positive impact on the treatment outcome, encouraging further trials on the strategy.

View this table:
  • View inline
  • View popup
Table 7—

Impact of granulocyte colony stimulating factor and dose intensity in treatment of small cell lung cancer

Extensive disease

Treatment outcome in extensive-stage SCLC is a sinister story. Various strategies have been investigated but progress has been minimal. With the introduction of etoposide and cisplatin in the late 1970s a new, active combination was available, partly noncross resistant to the CAV regime. A computer model by Goldie and Coldman 58 predicting superiority of alternating compared to sequential treatment with noncross resistant regimes, could thus be tested in clinical practice. The model was corroborated by a Canadian trial on 289 patients 14; while others were unable to prove statistically significant differences in survival, including a 3-armed intergroup trial on 437 patients 59. The principle does not add to either cost nor toxicity and it is still frequently applied in extensive disease.

Chute et al. 60 have made an interesting retrospective analysis of survival characteristics in extensive SCLC in North America in the last 20 yrs as reflected in data from the control arms of phase III trials and from cases registered in the Surveillance, Epidemiology, and End Results (SEER) database. Both data sources reflected an improvement in median as well as 5-yr survival when data from 1972–1981 were compared with data from 1982–1990. Median survival in trials increased from 7.0 to 8.9 months and from 6.5 to 8.2 months in the SEER database. The 5-yr survival rate rose from 0.8% to 1.6%. Least square regression analysis of data from the trials showed that treatment with cisplatin and year of study initiation were both significantly related to median survival time. The influence of “year of study initiation” could be caused by stage migration 61 but the SEER data suggested a prolongation for all SCLC patients over the period. Prolongation of survival in SCLC with time could, in addition to better combinations of cytotoxic agents, be a result of improved means of general medical management including specific supportive care.

The retrospective analysis cannot, however, point out especially beneficial treatment strategies for extensive SCLC. The clinical characteristics of these patients vary much more than in patients with limited disease; some only have a minor lesion such as a lymph node high on the neck while others have multiple metastases in several organs. Aiming to find a main thread Aisner 62 reviewed trials on extensive-stage SCLC from the 1990s. It was found that it was useful to categorize treatment strategies into one of three groups: The aggressive therapeutic approach (29 series), the intermediate regimes (10 series) and the minimal approach (for poor-prognosis and for elderly patients) (10 series). Complete remission (CR) rates were higher and median survival times longer in the aggressive category compared to those in the intermediate category, while data on other important end-points such as rates of early death and long-term survival and, not least, on quality of life (QoL) were rarely available. The “minimal” approach resulted in response and survival rates considerably below the figures in the other two groups. How were patients selected to treatment in one or the other category? Aggressive treatment is not for poor-risk patients. But what about the poor-risk patients? Is it clinically meaningful and ethically correct to operate with a minimal approach? Many in this category die early, because treatment is insufficient or because they are too sick when treatment is initiated. “An early death from progressive disease is not likely to improve QoL.” Aisner states 62, thinking of those who were undertreated, and continue “Whether the toxicity-to-benefit ratio is appropriate to an individual patient is therefore a matter to be discussed between the patient and his or her care provider.” This is a point, but Aisner 62 does not refer to a toxicity-to-benefit model which could help the patient and the caretaker with this difficult choice. Simple algorithms can be established, however, predicting risk of early death from pretreatment characteristics such as performance status, serum LDH and age 63. If performance status (PS)>1 and LDH above the upper normal limit each contribute with one risk point patients can be categorized into one of three risk groups as shown in table 8⇓. The data were retrieved from the Copenhagen database, supplemented with a European series 64 to illustrate the consistency of the “system”. With these two simple clinical attributes it is possible to allocate extensive-stage patients into risk groups with clinically important differences in survival. It is likely that 23% of the patients, who are in the best category, should receive aggressive treatment (and that some may benefit from additional chest irradiation) and that an intermediate treatment approach is optimal for group 2 patients. A meta-analysis on available trial data might answer the question. As for group 3, which includes as many as 40% of the patients, the treatment approach should probably be designed according to physical deficit: one for older patients, and others for patients with compromised lung or liver function, as for patients with a heavily infiltrated bone marrow. Some, desperately ill patients should not be treated at all. Trials aiming to find an appropriate treatment for poor-risk patients are rare.

View this table:
  • View inline
  • View popup
Table 8—

OlgpØy Hlrpn Uøle.a≤Uln f –ICU f3nro:rvdvlpdCØ eØCdyØoel, ØoslppcØppcC, clCcØr

An example is a feasibility study on per oral etoposide and cyclophosphamide in treatment of poor-prognosis extensive SCLC 65. Patients who had a low serum albumin or a poor performance status for entry in the current SWOG trial received the two agents for 14 days every 28 days, either once daily (18 patients) or twice daily (39 patients). Age of the patients ranged between 46–94 yrs, 81% had PS=2 (None had PS>2) and 35% had a serum albumin <3.5 g·dL−1. Grade 3–4 neutro- and leukopenia were observed in 58 and 53% of the patients, respectively. Early death or progression was observed in 37% of the patients, objective resonse was seen in 26% and unconfirmed response in further 16%. Median failure-free survival was 3–4 months. Serum etoposide on day 2 as well as pretreatment serum LDH were predicitive for the level of granulocyte nadir. Age was not included in the regression analysis. More appropriate regimes should be possible and further trials on this poorest category of the SCLC patients should be initiated.

Aggressive treatment approach for good-risk patients (patients aged <68 yrs and with a PS of 0–2) were investigated by Murray et al. 66. The regime: CODE, included weekly cisplatin and doxorubicine plus etoposide every second week. The control regime was CAV alternating with EP, three-times each. The response rate with CODE was higher (87 versus 69%) but progression-free and overall survival were not significantly better than in the control group. Death rates from neutropenic fever were 8% in the CODE arm versus 1% in the control arm. Ten per cent in both arms were progression free at 2 yrs. Thus, this trial did not support the idea that aggressive treatment is better than a conventional regime in good risk extensive SCLC.

Elderly patients

Elderly patients are, by convention, patients aged ≥70 yrs. In the 1970s many treatment trials did not include patients >70 yrs but most recent trials include patients aged 70–80 yrs. Compared to the incidence of lung cancer in this age group, however, these patients are underrepresented. This selection reflects a common clinical sense among lung physicians rather than in- and exclusion criteria for the individual trial. Elderly patients will often have cardiovascular disease, diabetes, reduced kidney function, locomotorical handicaps etc. making intensive chemo- and radiotherapy inappropriate. Gentle regimes for elderly patients have been investigated but simple guidelines and well-documented treatment options for making an easy choice are still a distant prospect. A retrospective clinical study of Dajczman et al. 67 from Quebec, Canada, gives a good impression of the clinical dilemma. Records were reviewed on 312 patients with a pathological diagnosis of SCLC at four McGill University Hospitals from a 7-yrs period: 1985–1991. Elderly patients (≥70 yrs) comprised 26% of the series, i.e. elderly patients were under-represented as there were 40% in this age group of SCLC patients notified in the British Columbia Cancer Registry in the same period 68). Staging and treatment of the elderly patients were compared to that of patients aged 60–69 yrs and <60 yrs, respectively. The treatment was regarded to have been “suboptimal” if neither chemo- or radiotherapy had been given to a patient with a performance status of ≤3. Suboptimal treatment was recorded in 23% of patients aged ≥70 yrs, compared to 9% of patients aged 60–69 yrs and 5% of patients aged <60 yrs. In limited stage patients chemo- plus radiotherapy was given to 43%, 65% and 69% of patients in the three age groups, respectively. There was no difference in proportions of limited stage disease: 43%, 40% and 45%, but elderly patients were less aggressively investigated compared to younger patients. Only one of 81 elderly patients was enrolled in an experimental protocol compared with 19 and 28% of the younger patient groups. Limited treatment of only 1–3 cycles of chemotherapy was quite frequent in elderly patients: 45% compared to 35% and 22% in the two younger age groups. Response and survival data in the treated patients reflected these differences in treatment intensity: CR or PR were seen in 25%, 49% and 41% in the three age categories; median survival figures were: 6, 9 and 8.5 months, respectively, but the 2-yr survival rates did not differ accordingly: being 8, 13 and 8%. Optimal versus suboptimal treatment was a significant and strong prognostic factor in a Cox analysis while survival was not affected by differences in patient age. These data on treatment outcome may support the view that choice and decisions made by the individual oncologist or lung physician seem to be reasonable. The larger proportions, however, of elderly patients who had few staging examinations, did not receive “optimal” therapy, did not receive chest radiation, only received “limited” chemotherapy, leaves the impression, that treatment outcome in these, selected, elderly patients could have been even better. In other words: a more active approach and uniform handling of elderly patients, including treatment regimens especially designed for this category of patients, are warranted. Ten-years-ago single-agent regimes with etoposide per orally were investigated as candidates for treatment of elderly SCLC patients 69–73. Given intraveneously in a dosage of 100 mg·m−2 for 5 days every 3 weeks Bork et al. 71 obtained CR or PR in 65% of the patients, the median survival was 10.6 months and 2-yr survival 8%. Per oral etoposide 200 mg·m−2 5 days every 3 weeks resulted in a similar response rate (62%), but both response duration and median survival were three months shorter. The 2-yr survival was 7% 73. Two trials comparing p.o. etoposide with i.v. combination chemotherapy have definitively changed the view on the p.o. etoposide regime. The British Medical Research Council conducted a trial on SCLC patients with poor performance (2–4), at any age. Median age was 67 yrs, range: 35–83 yrs. Patients were randomized to receive four-cycles of etoposide 50 mg twice daily for 10 days every 3 weeks or four cycles of a control regimen of intravenous EV or CAV. The trial was closed after inclusion of 339 patients (planned 450 patients) because of inferior results in the etoposide arm. Thus, >30% of the patients in this group died before the 3-month assessment. Median survival was 130 days compared to 183 days in patients treated with intravenous chemotherapy. The palliative effects of the treatments were similar. Same experience was obtained in a trial conducted at four London hospitals 74. Patients with extensive SCLC, performance status 0–3, were randomized to treatment with six-cycles of either p.o. etoposide 100 mg·m−2 for 5 days every 3 weeks or i.v. PE alternating with CAV. Median age of the patients was 66 yrs (50–86 yrs) and 67 yrs (49–80 yrs), respectively. The trial was stopped after inclusion of 155 patients from a projected intake of 365 patients because of inferior survival outcome in the etoposide arm. Median progression-free survival was only 3.6 months compared to 5.6 months in the control arm, 1-yr survival figures were 9.8% and 19.3%. Although the two trials did not specifically focus on the treatment of elderly patients, the outcome does not support continued use of the regimen in this group either.

A policy of combination chemotherapy regimes specifically designed for elderly patients has been investigated by the Canadian lung cancer group in Vancouver 68, 75. In the period 1982–1991 elderly or infirm patients with limited SCLC were offered a standard treatment of one-cycle CAV followed 3 weeks later by one-cycle EP plus concurrent thoracic radiotherapy, 20 Gy in 5 fractions or 30 Gy in 10 fractions. Prophylactic cranial irradiation was not routinely administered. The series included 55 patients, 67% of the patients were aged ≥70 yrs, 55% had PS 0–1, 71% had normal S-LDH, and 60% were females. Although the patients only received two-cycles of chemotherapy and a suboptimal radiotherapy, 28% of the patients survived 2 yrs and 18% 5 yrs.

In the following period: 1991–1994 the Vancouver group replaced the two series CAV with four series PAVE (cisplatin 30 mg·m−2 day 1, adriamycin 40 mg·m−2 day 1, vincristine 1 mg·m−2 day 1, etoposide 100 mg·m−2 days 1, 3, 5) plus the same simple irradiation programme for treatment of limited disease and in selected patients with extensive disease. The chest irradiation was given concomitantly with the second cycle of chemotherapy which was then reduced to only PE. A total of 66 patients were treated: 25 patients with limited disease, with a median age of 72 yrs (66–79 yrs), and 41 patients with extensive disease, median age 69 yrs (66–81 yrs). The 2-yr survival rates for limited and extensive stage patients were 38% and 18%, respectively, and the 5-yr survival rates were 24% and 5%. Prognostic attributes such as PS, LDH and metastatic sites did not suggest that these patients were especially selected. It is difficult to explain these favourable results on the basis of the general model of SCLC as a tumour with a high incidence of clonal heterogeneity but from a practical point of view the outcome is very stimulating for further development of specific treatment regimes for elderly patients. Two to four cycles of chemotherapy seem to be appropriate and chest irradiation can be given in less fractions, suggesting, that late lung toxicity does not seem to be a major problem in these patients.

A Japanese phase II trial 76 conducted in 1995–1996, also supports the supposition that a reasonably high initial dose level is important for the outcome. Thus, 36 patients, aged 73 yrs (70–80 yrs), received up to 4 cycles of carboplatin (AUC 5) plus etoposide 100 mg·m−2 i.v. days 1–3 every 4 weeks. The 44% of patients with limited disease had additional chest irradiation after chemotherapy. Sixty-one per cent of the patients received the scheduled 4 cycles and dose reductions were only necessary in 10% of the patients. Median survival was 10.8 months and 1 and 2-yr survival rates were 47.2% and 15.4%, respectively. Serum LDH, stage and anaemia were significant prognostic factors in a Cox analysis but age was not.

The high rates of long-term survival in these patients could hardly have been obtained without chest irradiation. A subgroup analysis in the meta analysis on thoracic radiotherapy for SCLC 2 proved no benefit of chest irradiation in patients aged ≥70 yrs, but these data derived from trials using other agents and other radiation schedules such as split course and greater doses on fewer fractions, resulting in greater toxicity than seen today. In current regimes elderly people seem to both tolerate and to benefit from chest irradiation. As an example, the intergroup trial comparing twice-daily with once-daily thoracic radiotherapy 3 patients aged 82 yrs were included. Outcome was independent of age 77 and age was not a risk factor in a multivariable Cox analysis 3. Treatment in these series also included prophylactic-cranial irradiation to patients in CR and no extraordinary neurotoxicity was observed. Systematic assessments, however, in larger series would be useful. Finally, the populations are changing and the increasing number of elderly people in good general health represents both a stimulus and a demand for development of specific regimes for elderly patients. Many elderly will tolerate combination chemotherapy and all will expect to be offered an individualized, documented and efficacious treatment.

New chemotherapeutic agents

The activity of six new agents have been investigated in phase I, II and III trials during the past 9 yrs but the definitive roles of each agent have not yet been defined 78, 79. The agents are two taxanes: paclitaxel and docetaxel, two topoisomerase I inhibitors: topotecan and irinotecan, an antimetabolite: gemcitabine and a spindle inhibitor: vinorelbine 80–86 (table 9⇓) (a seventh agent: oxaliplatin, has not yet been tested in SCLC). The phase II trials include previously untreated patients with extensive disease or patients with relapse after >3 months in remission after first-line treatment. A few trials include patients with refractory tumours on first-line treatment. Some trials use G-CSF as rescue or as an integrated part of the treatment. The addition of G-CSF has an influence on the observed haematological toxicity and may have an impact on the response rates. The strategy thus makes it more difficult to compare the outcome with that from other trials.

View this table:
  • View inline
  • View popup
Table 9—

OlgpØy H.loØ enØlesØCerdlpd7de. CØ71, ØCeo

Comparison of haematological toxiciy data is furthermore difficult because of various ways to describe the observations: nadirs in first cycle or global nadir from all cycles; per cent of patients or per cent of series with grade 3 or grade 4 toxicity. Survival data should be interpreted cautiously because similar pretreatment characteristics of the patients do not prove the same degree of selection.

The taxanes

Two trials investigated the activity of paclitaxel 250 mg·m−2 by 24-h infusion 80, 81 (table 9⇑) and achieved response rates as high as 34% and 35%, respectively. The 24-h infusions have since been replaced by 3-h or 1-h infusion times. The high response rates have stimulated several trials with paclitaxel in combination with cisplatin, carboplatin, and etoposide 87–93 (table 10⇓). Investigations on paclitaxel added to the “standard” regimen of cisplatin plus etoposide has naturally had a high priority. Appropriate doses of the three agents have been clarified in a phase I trial in extensive stage patients 90, recommending paclitaxel 175 mg·m−2 over a 3-h period, cisplatin 80 mg·m−2 and etoposide 80 mg·m−2 i.v. on day 1 and 160 mg·m−2 p.o. on days 2–3. G-CSF was only used according to the American Society of Clinical Oncologists' guidelines. The regimen was subsequently investigated by SWOG in a phase II trial on 90 patients with extensive disease 91. A median survival of 11 months seems encouraging and 7% toxic deaths and nearly 40% with grade 4 neutropenia is not considered extraordinary for a series of patients with extensive SCLC.

View this table:
  • View inline
  • View popup
Table 10—

OlgpØDy H.lo Ø erdlpo7de.ellCØo

Glisson et al. 89 gave paclitaxel 130 mg·m−2 over 3 h, cisplatin 75 mg·m−2 on day 1, and etoposide 80 mg·m−2 i.v. on days 1–3 in a trial on 41 patients. Two early septic deaths were observed and grade 4 neutropenia was observed in 44% of 188 courses. The response rate was 90%. A Greek randomized trial 92 including 45% patients with limited disease was stopped prematurely after inclusion of 133 patients because of eight toxic deaths in the three-agents arm compared to none among patients treated with cisplatin and etoposide alone. Same doses as in the SWOG trial were used, G-CSF was used prophylacticly. There was no significant difference in survival between the arms. Grade 4 neutropenia occurred in 39% of the patients, i.e. as in the phase I and II trials. The toxicity problem must thus be taken serious. Poor-risk patients should be excluded and conservative dose reduction rules may be a safer policy than giving G-CSF on demand.

Carboplatin plus etoposide with or without paclitaxel (4 cycles) is under investigation in a randomized trial now including 170 patients (Birch et al. 93). Patients with limited disease (53%) receive concurrent chest radiation with cycles 3 and 4. No extraordinary toxicity is mentioned. The response rates in the paclitaxel arm are modestly higher and there is a trend toward improved survival in patients with extensive disease.

Many trials with paclitaxel are still ongoing and it is too early to assess the role of this agent. Other schedules of paclitaxel are under investigation 79, e.g. day 10 after cisplatin, as a 24-h infusion on day 5 after topotecan on days 1–5, day 10 after etoposide twice daily days 1–10, or followed by oral etoposide days 2–8. Currently no specific use of paclitaxel in the treatment of SCLC can be advocated, and it is still too early to guess which role paclitaxel will come to play in the treatment of SCLC.

Docetaxel has been much less investigated and the outcome data is not especially promising. A phase II trial on 43 previously untreated patients with extensive disease 82 resulted in a response rate of only 23%. The dose was 100 mg·m−2 every 3 weeks. Docetaxel plus doxorubicin in extensive SCLC were investigated in a phase II trial which was discontinued after inclusion of 20 patients because only two patients achieved a PR 94 (table 10⇑). A much better outcome was obtained when combined with cisplatin 95 and it is still too early to preclude activity of docetaxel in treatment of SCLC.

The topoisomerase I inhibitors

The topoisomerase II inhibitor etoposide is fundamental in the treatment of SCLC and another key role is expected for one or both of the two type I inhibitors: topotecan and irinotecan. The sensitivity patterns of the two agents are much alike when tested on cell lines 60 but different from that of etoposide. Both drugs are active in phase II trials on chemo-naïve patients and on patients with previously sensitive tumours 96–100 (table 11⇓). In a phase II trial on 48 chemo-naïve patients with extensive disease 96 topotecan, 2.0 mg·m−2 i.v. days 1–5 every 3 weeks, resulted in an objective response rate of 39%, median response duration was 4.8 months and median survival 10 months. Prophylactic G-CSF was given to the last 35 patients, reducing occurrence of grade 3–4 haematological toxicity from 92% to 29% of the patients. Currently recommended dose is 1.5 mg·m−2 i.v. for 5 days every 3 weeks 101. The agent is well absorbed and the efficacy of tablet treatment is under evaluation in large randomized trials performed by the producer. Single-agent treatment with irinotecan 60 mg·m−2 weekly×3 in 4 week cycles results in similar response rates 102 and response rates as high as 84% were seen when the agent was combined with cisplatin (60 mg·m−2 every 4 weeks) in 75 chemo-naïve patients 98 and 71% when combined with etoposide 99 as second-line treatment. These two combinations are especially interesting: firstly, is a topoisomerase I inhibitor plus cisplatin as efficacious or even better than etoposide plus platinum? And secondly, will alternating (or combined) dosage of a topo I and a topo II agent delay upregulation of topoisomerase 1/2 activity in the tumour cells and thus reduce the risk of treatment resistance?

View this table:
  • View inline
  • View popup
Table 11—

Phase II/III trials with topoisomerase I inhibitors

Question number 1 has already been investigated in a randomized trial on extensive disease comparing irinotecan plus cisplatin (IP) with etoposide plus cisplatin (EP) 100. The trial was halted at an interims analysis after inclusion of 154 patients because of major difference in survival: the median survival in the IP arm was 420 days compared to 300 days in the EP arm (p=0.0047) and 1-yr survival 60% versus 40%. Only 27% of patients receiving IP had grade 3–4 neutropenia compared to 52% in the EP arm. More definitive statements about efficacy of the IP regimen must await conclusion of the trial plus a confirming trial.

Introduction of topoisomerase inhibitors in treatment of limited-stage disease is under way, but there is some anxiety about the toxicity if given concurrently with radiotherapy 103. Both agents have been investigated in phase I trials on nonsmall cell lung cancer 104, 105. Severe cases of oesophagitis and pneumonitis were observed in both trials and dose reductions to 33–50% of the otherwise recommended doses seem to be necessary for both agents.

Gemcitabine

Gemcitabine is an antimetabolite and it has a sensitivity pattern which differs from that of other active agents in SCLC. The haematological toxicity of the drug is modest. The agent has proved activity in phase II trials (table 9⇑) but data on its activity in combination regimens is restricted. The Italian “Lazio” group 106 added gemcitabine to the EP regimen in treatment of 43 patients, limited as well as extensive stage disease. The overall response rate was 68% with CR rates of 24% in limited stage and 0% in extensive stage. Dosage of etoposide was only 50 mg·m−2 i.v. day 3–5 and grade 3–4 neutropenia was seen in only 29% of the cycles reflecting that gemcitabine can be included in a more dose-intensive regime.

Vinorelbine

Vinorelbine is a semisynthetic vinca alkaloid like vincristine and vindesine, which are both active in SCLC. The activity of vinorelbine in phase II trials has been modest (table 9⇑) but response rates of up to 70% have been obtained in combination with cisplatin and etoposide and with carboplatin 79. The sensitivity profile of the vinca alkaloids and the taxanes are much alike 107 and the clinical activity of vinorelbine in relation to that of vincristine and of paclitaxel should be investigated further in treatment trials.

Concluding remarks

How can these new agents best be integrated in combination regimens for the treatment of SCLC? Are the design of clinical trials sufficiently systematic? Are there too many phase II and too few randomized trials? Should more attention be paid on laboratory data such as sensitivity profiles in cell lines 107 before new combinations of new and old agents are investigated in the clinic? A majority of the trials on the new agents have been supported by the drug companies. Clinicians and drug companies have, in the early phase, a common interest in defining a role for the individual agent. Then ways often part. New agents get ready from the “pipelines”, phase I and II trials are requested and it may then be difficult for the clinicians to find time and resources for large, investigator initiated, randomized trials. And randomized trials must be large, typically 250–300 patients in each arm in order to gain sufficient statistical power. To add insult to injury requirements to documentation have been strengthened, it is hardly possible today to have a protocol approved by the scientific ethical committee if it does not fulfil the good clinical practice (GCP) criteria.

It is important to realize and to tell both the public and health authorities that there is not yet a really efficacious standard treatment for SCLC. Patients with SCLC must still be ready to be treated in trials and new (expensive) agents should be restricted to investigative regimes for the sake of continued clinical research and to secure that the lions share of the drug budget goes to development.

  • Received July 31, 2001.
  • Accepted August 20, 2001.
  • © ERS Journals Ltd

References

  1. ↵
    Lassen U, Østerlind K, Hansen M, Dombernowsky P, Bergman B, Hansen HH. Long-term survival in small-cell lung cancer: Posttreatment characteristics in patients surviving 5 to 18+ years. An analysis of 1,714 consecutive patients. J Clin Oncol 1995;13:1215–1220.
    OpenUrlAbstract
  2. ↵
    Pignon JP, Arriagada R, Ihde DC, et al. A meta-analysis of thoracic radiotherapy for small cell lung cancer. N Engl J Med 1992;327:1618–1624.
    OpenUrlPubMedWeb of Science
  3. ↵
    Turrisi AT III, Kim K, Blum R, et al. Twice-daily compared with once-daily thoracic radiotherapy in limited small-cell lung cancer treated concurrently with cisplatin and etoposide. N Engl J Med 1999;340:265–271.
    OpenUrlCrossRefPubMedWeb of Science
  4. ↵
    Hansen HH, Dombernowsky P, Hirsch FR, Hansen M, Rygard J. Prophylactic irradiation in bronchogenic small cell anaplastic carcinoma. A comparative trial of localized versus extensive radiotherapy including prophylactic brain irradiation in patients receiving combination chemotherapy. Cancer 1980;46:279–284.
    OpenUrlCrossRefPubMedWeb of Science
  5. ↵
    Aupérin A, Arriagada R, Pignon J-P, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. N Engl J Med 1999;341:476–484.
    OpenUrlCrossRefPubMedWeb of Science
  6. ↵
    Rawson NSB, Peto J. An overview of prognostic factors in small cell lung cancer. A report from the subcommittee for the management of lung cancer of the United Kingdom coordinating committee on cancer research. Br J Cancer 1990;61:597–604.
    OpenUrlPubMedWeb of Science
  7. Østerlind K, Andersen PK. Prognostic factors in small cell lung cancer: Multivariate model based on 778 patients treated with chemotherapy with or without irradiation. Cancer Res 1986;46:4189–4194.
    OpenUrlAbstract/FREE Full Text
  8. Souhami RL, Bradbury I, Geddes DM, Spiro SG, Harper PG, Tobias JS. Prognostic significance of laboratory parameters measured at diagnosis in small cell carcinoma of the lung. Cancer Res 1985;45:2878–2882.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Vincent MD, Ashley SE, Smith IE. Prognostic factors in small cell lung cancer: A simple prognostic index is better than conventional staging. Eur J Cancer Clin Oncol 1987;23:1589–1599.
    OpenUrlCrossRefPubMedWeb of Science
  10. ↵
    Lassen U, Hansen HH. Surgery in limited stage small cell lung cancer. Cancer Treat Rev 1999;25:67–72.
    OpenUrlCrossRefPubMedWeb of Science
  11. ↵
    De Leyn P, Vansteenkiste J, Cuypers P, et al. Role of cervical mediastinoscopy in staging of non-small cell lung cancer without enlarged mediastinal lymph nodes on CT scan. Eur J Cardio-thorac Surg 1997;12:706–712.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Østerlind K, Hansen M, Hansen HH, Dombernowsky P, Rørth M. Treatment policy of surgery in small cell carcinoma of the lung: Retrospective analysis of a series of 874 consecutive patients. Thorax 1985;40:272–277.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Goldie JH, Coldman AJ, Gudauskas GA. Rationale for the use of alternating non-cross-resistant chemotherapy. Cancer Treat Rep 1982;66:439–449.
    OpenUrlPubMedWeb of Science
  14. ↵
    Evans WK, Feld R, Murray N, et al. Superiority of alternating non-cross-resistant chemotherapy in extensive small cell lung cancer. A multicenter, randomized clinical trial by the National Cancer Institute of Canada. Ann Int Med 1987;107:451–458.
  15. Feld R, Evans WK, Coy P, et al. Canadian multicenter randomized trial comparing sequential and alternating administration of two non-cross-resistant chemotherapy combinations in patients with limited small-cell carcinoma of the lung. J Clin Oncol 1987;5:1401–1409.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Østerlind K. Alternating or sequential chemotherapy in small cell lung cancer? Lung Cancer 1989;5:173–177.
    OpenUrlCrossRef
  17. ↵
    Arriagada R, Le Chevalier T, Borie F, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. J Natl Cancer Inst 1995;87:183–190.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Warde P, Payne D. Does thoracic irradiation improve survival and local control in limited-stage small cell carcinoma of the lung? A meta-analysis. J Clin Oncol 1992;10:890–895.
    OpenUrlAbstract
  19. ↵
    Paesmans M, Mascaux C, Berghmans T, et al. Etoposide and cisplatin merit their key role in chemotherapy for small cell lung cancer: A meta-analysis with a methodology assessment, by the European Lung Cancer Working Party (ELCWP). Proceedings of ASCO 18. 1999.
  20. ↵
    Murray N, Coy P, Pater JL, et al. Importance of timing for thoracic irradiation in the combined modality treatment of limited-stage small-cell lung cancer. J Clin Oncol 1993;11:336–344.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Work E, Nielsen OS, Bentzen SM, Fode K, Palshof T. Randomized study of initial versus late chest irradiation combined with chemotherapy in limited-stage small-cell lung cancer. J Clin Oncol 1997;15:3030–3037.
    OpenUrlAbstract
  22. ↵
    Gregor A, Cull A, Stephens RJ, et al. Prophylactic cranial irradiation is indicated following complete response to induction therapy in small cell lung cancer: results of a multicentre randomised trial. United Kingdom Coordinating Committee for Cancer Research (UKCCCR) and the European Organization for Research and Treatment of Cancer (EORTC). Eur J Cancer 1997;33:1752–1758.
  23. ↵
    Johnson DH, Turrisi AT, Chang AY, et al. Alternating chemotherapy and twice-daily thoracic radiotherapy in limited-stage small-cell lung cancer: A pilot study of the Eastern Cooperative Oncology Group. J Clin Oncol 1993;11:879–884.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Johnson DH, Bass D, Einhorn LH, et al. Combination chemotherapy with or without thoracic radiotherapy in limited-stage small-cell lung cancer: A randomized trial of the Southeastern Cancer Study Group. J Clin Oncol 1993;11:1223–1229.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S. Initial versus delayed accelerated hyperfractionated radiation therapy and concurrent chemotherapy in limited small-cell lung cancer: a randomized study. J Clin Oncol 1997;15:893–900.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Bonner JA, Sloan JA, Shanahan TG, et al. Phase III comparison of twice-daily split-course irradiation versus once-daily irradiation for patients with limited stage small-cell lung carcinoma. J Clin Oncol 1999;17:2681–2691.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Mennecier B, Jacoulet P, Dubiez A, et al. Concurrent cisplatin/etoposide chemotherapy plus twice daily thoracic radiotherapy in limited stage small cell lung cancer: a phase II study. Lung Cancer 2000;27:137–143.
    OpenUrlCrossRefPubMedWeb of Science
  28. ↵
    Nishiwaki Y, Fukuoka M, Yokoyama A, et al. Phase III study of concurrent versus sequential thoracic radiotherapy in combination with cisplatin and etoposide for limited-stage small cell lung cancer: The Japan Clinical Oncology Group study. Lung Cancer 2000;29:Suppl. 2, 163.
  29. ↵
    Choi NC, Herndon JE, Rosenman J, et al. Phase I study to determine the maximum-tolerated dose of radiation in standard daily and hyperfractionated-accelerated twice-daily radiation schedules with concurrent chemotherapy for limited-stage small-cell lung cancer. J Clin Oncol 1998;16:3528–3536.
    OpenUrlAbstract
  30. ↵
    Kristjansen PEG, Hansen HH. Brain metastases from small cell lung cancer treated with combination chemotherapy. Eur J Cancer Clin Oncol 1988;24:545–549.
    OpenUrlCrossRefPubMedWeb of Science
  31. ↵
    Pedersen AG. Diagnostic procedures in the detection of CNS metastases from small cell lung cancer In: Hansen HH, ed. Lung cancer: Basic and clinical aspectsBoston, Martinus Nijhoff Publishers, 1986; pp. 153–182.
  32. ↵
    Bach F, Sorensen JB, Adrian L, et al. Brain relapses in chemotherapy-treated small cell lung cancer: a retrospective review of two time-dose regimens of therapeutic brain irradiation. Lung Cancer 1996;15:171–181.
    OpenUrlCrossRefPubMedWeb of Science
  33. ↵
    Laplanche A, Monnet I, Santos-Miranda JA, et al. Controlled clinical trial of prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. Lung Cancer 1998;21:193–201.
    OpenUrlCrossRefPubMedWeb of Science
  34. ↵
    Carney DN. Prophylactic cranial irradiation and small-cell lung cancer (Editorial). N Engl J Med 1999;341:524–526.
    OpenUrlCrossRefPubMedWeb of Science
  35. ↵
    Meert AP, Paesmans M, Berghmans T, et al. Prophylactic cranial irradiation in small cell lung cancer: a systematic review of the literature with meta-analysis. BMC Cancer 2001;1:5.
    OpenUrlCrossRefPubMed
  36. ↵
    The Prophylactic Cranial Irradiation Overview Collaborative Group. Cranial irradiation for preventing brain metastases of small cell lung cancer in patients in complete remission (Cochrane Review). The Cochrane Library (2). 2001;. Oxford. Update Software.
  37. ↵
    Johnson BE, Patronas N, Hayes W, et al. Neurologic, computed cranial tomographic, and magnetic resonance imaging abnormalities in patients with small-cell lung cancer: Further follow-up of 6- to 13-year survivors. J Clin Oncol 1990;8:48–56.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Komaki R, Meyers CA, Shin DM, et al. Evaluation of cognitive function in patients with limited small cell lung cancer prior to and shortly following prophylactic cranial irradiation. Int J Radiat Oncol Biol Phys 1995;33:179–182.
    OpenUrlCrossRefPubMedWeb of Science
  39. ↵
    Souhami RL, Harper PG, Linch D, et al. High-dose cyclophosphamide with autologous marrow transplantation as initial treatment of small cell carcinoma of the bronchus. Cancer Chemother Pharmacol 1982;8:31–34.
    OpenUrlCrossRefPubMedWeb of Science
  40. ↵
    Spitzer G, Farha P, Valdivieso M, et al. High-dose intensification therapy with autologous bone marrow support for limited small-cell bronchogenic carcinoma. J Clin Oncol 1986;4:4–13.
    OpenUrlAbstract
  41. ↵
    Klasa RJ, Murray N, Coldman AJ. Dose-intensity meta-analysis of chemotherapy regimens in small-cell carcinoma of the lung. J Clin Oncol 1991;9:499–508.
    OpenUrlAbstract
  42. ↵
    Brugger W, Frommhold H, Pressler K, Mertelsmann R, Kanz L. Use of high-dose etoposide/ifosfamide/carboplatin/epirubicin and peripheral blood progenitor cell transplantation in limited-disease small cell lung cancer. Semin Oncol 1995;22:3–8.
    OpenUrlPubMedWeb of Science
  43. ↵
    Elias AD, Ayash L, Frei E III, et al. Intensive combined modality therapy for limited-stage small-cell lung cancer. J Natl Cancer Inst 1993;85:559–566.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    van der Velde, Bosquee L, Weynants P, Canon JL, Rosier JF, Humblet Y. Moderate dose-escalation of combination chemotherapy with concomitant thoracic radiotherapy in limited-disease small-cell lung cancer: prolonged intrathoracic tumor control and high central nervous system relapse rate. Groupe d'Oncologie-Pneumologie Clinique de l'Universite Catholique de Louvain, Brussels and Liege, Belgium. Ann Oncol 1999;10:1051–1057.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Leyvraz S, Perey L, Rosti G, et al. Multiple courses of high-dose ifosfamide, carboplatin, and etoposide with peripheral-blood progenitor cells and filgrastim for small-cell lung cancer: A feasibility study by the European Group for Blood and Marrow Transplantation. J Clin Oncol 1999;17:3531–3539.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Bessho A, Ueoka H, Kiura K, et al. High-dose ifosfamide, carboplatin and etoposide with autologous peripheral blood progenitor cell transplantation for small-cell lung cancer. Anticancer Res 1999;19:693–698.
    OpenUrlPubMedWeb of Science
  47. ↵
    Crawford J, Ozer H, Stoller R, et al. Reduction by granulocyte colony-stimulating factor of fever and neutropenia induced by chemotherapy in patients with small-cell lung cancer. N Engl J Med 1991;325:164–170.
    OpenUrlCrossRefPubMedWeb of Science
  48. ↵
    Trillet-Lenoir V, Green J, Manegold C, et al. Recombinant granulocyte colony stimulating factor reduces the infectious complications of cytotoxic chemotherapy. Eur J Cancer 1993;29A:319–324.
  49. ↵
    Nichols CR, Fox EP, Roth BJ, Williams SD, Loehrer PJ, Einhorn LH. Incidence of neutropenic fever in patients treated with standard-dose combination chemotherapy for small-cell lung cancer and the cost impact of treatment with granulocyte colony-stimulating factor. J Clin Oncol 1994;12:1245–1250.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Radford JA, Ryder WDJ, Dodwell D, Anderson H, Thatcher N. Predicting septic complications of chemotherapy: an analysis of 382 patients treated for small cell lung cancer without dose reduction after major sepsis. Eur J Cancer 1993;29A:81–86.
    OpenUrlCrossRef
  51. ↵
    Østerlind K, Pedersen AG, Dombernowsky P, Hansen M, Hansen HH. Predictability of bone marrow toxicity in chemotherapy of small cell lung cancer. Lung Cancer 1994;11:91.
  52. ↵
    Bunn PA Jr, Crowley J, Kelly K, et al. Chemoradiotherapy with or without granulocyte-macrophage colony-stimulating factor in the treatment of limited-stage small-cell lung cancer: a prospective phase III randomized study of the Southwest Oncology Group [published erratum appears in J Clin Oncol 1995 Nov; 13(11): 2860]. J Clin Oncol 1995;13:1632–1641.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Pujol JL, Douillard JY, Riviere A, et al. Dose-intensity of a four-drug chemotherapy regimen with or without recombinant human granulocyte-macrophage colony-stimulating factor in extensive-stage small-cell lung cancer: a multicenter randomized phase III study. J Clin Oncol 1997;15:2082–2089.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Steward WP, Von Pawel J, Gatzemeier U, et al. Effects of granulocyte-macrophage colony-stimulating factor and dose intensification of V-ICE chemotherapy in small-cell lung cancer: a prospective randomized study of 300 patients. J Clin Oncol 1998;16:642–650.
    OpenUrlAbstract
  55. ↵
    Woll PJ, Hodgetts J, Lomax L, Bildet F, Cour-Chabernaud V, Thatcher N. Can cytotoxic dose-intensity be increased by using granulocyte colony-stimulating factor? A randomized controlled trial of lenograstim in small-cell lung cancer. J Clin Oncol 1995;13:652–659.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Furuse K, Fukuoka M, Nishiwaki Y, et al. Phase III study of intensive weekly chemotherapy with recombinant human granulocyte colony-stimulating factor versus standard chemotherapy in extensive-disease small-cell lung cancer. The Japan Clinical Oncology Group. J Clin Oncol 1998;16:2126–2132.
    OpenUrlAbstract
  57. ↵
    Thatcher N, Girling DJ, Hopwood P, Sambrook RJ, Qian W, Stephens RJ. Improving survival without reducing quality of life in small-cell lung cancer patients by increasing the dose-intensity of chemotherapy with granulocyte colony-stimulating factor support: results of a British Medical Research Council Multicenter Randomized Trial. Medical Research Council Lung Cancer Working Party. J Clin Oncol 2000;18:395–404.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Goldie JH, Coldman AJ. Quantitative model for multiple levels of drug resistance in clinical tumors. Cancer Treat Rep 1983;67:923–931.
    OpenUrlPubMedWeb of Science
  59. ↵
    Roth BJ, Johnson DH, Einhorn LH, et al. Randomized study of cyclophosphamide, doxorubicin, and vincristine versus etoposide and cisplatin versus alternation of these two regimens in extensive small-cell lung cancer: A phase III trial of the Southeastern Cancer Study Group. J Clin Oncol 1992;10:282–291.
    OpenUrlAbstract
  60. ↵
    Chute JP, Chen T, Feigal E, Simon R, Johnson BE. Twenty years of phase III trials for patients with extensive-stage small-cell lung cancer: perceptible progress. J Clin Oncol 1999;17:1794–1801.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Feinstein AR, Sosin DM, Wells CK. The Will Rogers phenomenon. Stage migration and new diagnostic techniques as a source of misleading statistics for survival in cancer. N Engl J Med 1985;312:1604–1608.
    OpenUrlCrossRefPubMedWeb of Science
  62. ↵
    Aisner J. Extensive-disease small-cell lung cancer: the thrill of victory; the agony of defeat. J Clin Oncol 1996;14:658–665.
    OpenUrlFREE Full Text
  63. ↵
    Lassen UN, Osterlind K, Hirsch FR, Bergman B, Dombernowsky P, Hansen HH. Early death during chemotherapy in patients with small-cell lung cancer: derivation of a prognostic index for toxic death and progression. Br J Cancer 1999;79:515–519.
    OpenUrlCrossRefPubMedWeb of Science
  64. ↵
    Jørgensen LGM, Østerlind K, Genollá J, et al. Serum neuron-specific enolase (S-NSE) and the prognosis in small-cell lung cancer (SCLC): a combined multivariable analysis on data from nine centres. Br J Cancer 1996;74:463–467.
    OpenUrlPubMedWeb of Science
  65. ↵
    Grunberg SM, Crowley J, Hande KR, et al. Treatment of poor-prognosis extensive disease small-cell lung cancer with an all-oral regimen of etoposide and cyclophospham. Cancer Chemother Pharmacol 1999;44:461–468.
    OpenUrlCrossRefPubMedWeb of Science
  66. ↵
    Murray N, Livingston RB, Shepherd FA, et al. Randomized study of CODE versus alternating CAV/EP for extensive-stage small-cell lung cancer: an Intergroup Study of the National Cancer Institute of Canada Clinical Trials Group and the Southwest Oncology Group. J Clin Oncol 1999;17:2300–2308.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Dajczman E, Fu LY, Small D, Wolkove N, Kreisman H. Treatment of small cell lung carcinoma in the elderly. Cancer 1996;77:2032–2038.
    OpenUrlCrossRefPubMedWeb of Science
  68. ↵
    Murray N, Grafton C, Shah A, et al. Abbreviated treatment for elderly, infirm, or noncompliant patients with limited-stage small-cell lung cancer. J Clin Oncol 1998;16:3323–3328.
    OpenUrlAbstract
  69. ↵
    Clark PI, Cottier B. The activity of 10-, 14-, and 21-day schedules of single-agent etoposide in previously untreated patients with extensive small cell lung cancer. Semin Oncol 1992;19:36–39.
    OpenUrl
  70. Bork E, Ersbøll J, Dombernowsky P, Bergman B, Hansen M, Hansen HH. Teniposide and etoposide in previously untreated small-cell lung cancer: A randomized study. J Clin Oncol 1991;9:1627–1631.
    OpenUrlAbstract
  71. ↵
    Bork E, Sigsgaard T, Nissen KM, et al. Etoposide and teniposide in untreated small cell lung cancer. Lung Cancer 1991;7:Suppl., 121.
  72. Smit EF, Carney DN, Harford P, Sleijfer DT, Postmus PE. A phase II study of oral etoposide in elderly patients with small cell lung cancer. Thorax 1989;44:631–633.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    Bork E, Hirsch FR, Jeppesen N, Lassen U, Vallentin S, Østerlind K. Oral etoposide (VP-16) every 3 wks. or continuously to elderly patients with small cell lung cancer (SCLC): Preliminary results of a randomised study. Lung Cancer 1997;18:25.
    OpenUrl
  74. ↵
    Souhami RL, Spiro SG, Rudd RM, et al. Five-day oral etoposide treatment for advanced small-cell lung cancer: randomized comparison with intravenous chemotherapy. J Natl Cancer Inst 1997;89:577–580.
    OpenUrlFREE Full Text
  75. ↵
    Westeel V, Murray N, Gelmon K, et al. New combination of the old drugs for elderly patients with small-cell lung cancer: a phase II study of the PAVE regimen. J Clin Oncol 1998;16:1940–1947.
    OpenUrlAbstract
  76. ↵
    Okamoto H, Watanabe K, Nishiwaki Y, et al. Phase II study of area under the plasma-concentration-versus-time curve-based carboplatin plus standard-dose intravenous etoposide in elderly patients with small-cell lung cancer. J Clin Oncol 1999;17:3540–3545.
    OpenUrlAbstract/FREE Full Text
  77. ↵
    Yuen A, Zou G, Turrisi A, et al. Similar outcome of elderly patients in Intergroup trial 0096: Cisplatin, etoposdie, and thoracic radiotherapy administered once or twice daily in limited stage small cell lung cancer. Proceedings of ASCO 18. 1999.
  78. ↵
    Argiris A, Murren JR. Advances in chemotherapy for small cell lung cancer: single-agent activity of newer agents. Cancer J 2001;7:228–235.
    OpenUrlPubMedWeb of Science
  79. ↵
    Kelly K. New chemotherapy agents for small cell lung cancer. Chest 2000;117:156S–162S.
    OpenUrlCrossRefPubMedWeb of Science
  80. ↵
    Ettinger DS, Finkelstein DM, Sarma RP, Johnson DH. Phase II study of paclitaxel in patients with extensive-disease small-cell lung cancer: an Eastern Cooperative Oncology Group study. J Clin Oncol 1995;13:1430–1435.
    OpenUrlAbstract
  81. ↵
    Kirschling RJ, Grill JP, Marks RS, et al. Paclitaxel and G-CSF in previously untreated patients with extensive stage small-cell lung cancer: a phase II study of the North Central Cancer Treatment Group. Am J Clin Oncol 1999;22:517–522.
    OpenUrlCrossRefPubMedWeb of Science
  82. ↵
    Hesketh PJ, Crowley JJ, Burris HA III, et al. Evaluation of docetaxel in previously untreated extensive-stage small cell lung cancer: a Southwest Oncology Group phase II trial. Cancer J Sci Am 1999;5:237–241.
    OpenUrlPubMedWeb of Science
  83. ↵
    Huber RM, Gatzemeier U, Gosse H, et al. Topotecan in second-line therapy of SCLC: Impact on survival? Onkologie 2000;23:9–12.
  84. Cormier Y, Eisenhauer E, Muldal A, et al. Gemcitabine is an active new agent in previously untreated extensive small cell lung cancer (SCLC). A study of the National Cancer Institute of Canada Clinical Trials Group. Ann Oncol 1994;5:283–285.
    OpenUrlAbstract/FREE Full Text
  85. Postmus PE, Schramel FM, Smit EF. Evaluation of new drugs in small cell lung cancer: the activity of gemcitabine. Semin Oncol 1998;25:79–82.
    OpenUrlPubMedWeb of Science
  86. ↵
    Masters GA, Declerck L, Blanke CD, et al. Gemcitabine in refractory or relapsed small cell lung cancer (SCLC) - E1597: An ECOG phase II trial. Proceedings of ASCO 19, 527a. 2000.
  87. ↵
    Nair S, Marschek R, Grill J. A phase II study of paclitaxel (Taxol) and cisplatin (CDDP) in the treatment of extensive stage small cell lung cancer. Proceedings of ASCO 16, 454a. 1997.
  88. ↵
    Groen HJ, Fokkema E, Biesma B, et al. Paclitaxel and carboplatin in the treatment of small-cell lung cancer patients resistant to cyclophosphamide, doxorubicin, and etoposide: a non-cross-resistant schedule. J Clin Oncol 1999;17:927–932.
    OpenUrlAbstract/FREE Full Text
  89. ↵
    Glisson BS, Kurie JM, Perez-Soler R, et al. Cisplatin, etoposide, and paclitaxel in the treatment of patients with extensive small-cell lung carcinoma. J Clin Oncol 1999;17:2309–2315.
    OpenUrlAbstract/FREE Full Text
  90. ↵
    Kelly K, Pan Z, Wood ME, Murphy J, Bunn PA Jr. A phase I study of paclitaxel, etoposide, and cisplatin in extensive stage small cell lung cancer. Clin Cancer Res 1999;5:3419–3424.
    OpenUrlAbstract/FREE Full Text
  91. ↵
    Kelly K, Bunn PA, Lovato L, Crowley J, Livingston RB, Gandara DR. Final results fromSouthwest Oncology Group trial (SWOG) 9705: A phase II trial of cisplatin, etoposide and paclitaxel (PET) with G-CSF in untreated patients with extensive stage small cell lung cancer (SCLC). Proceedings of ASCO 19, 492a. 2000.
  92. ↵
    Mavroudis D, Papadakis E, Veslemes M, et al. A multicenter randomized phase III study comparing paclitaxel-cisplatin-etoposide (TEP) versus cisplatin-etoposide (EP) as front-line treatment in patients with small cell lung cancer (SCLC). Proceedings of ASCO 19, 484a. 2000.
  93. ↵
    Birch R, Greco FA, Hainsworth JD, et al. Preliminary results of a randomized study comparing etoposide and carboplatin (EP) with or without paclitaxel (T) in newly diagnosed small cell lung cancer. Proceedings of ASCO 19, 490a. 2000.
  94. ↵
    Ferri W. Phase II trial of docetaxel and doxorubicin in patients with extensive stage small cell lung cancer. Proceedings of ASCO 19, 540a. 2000.
  95. ↵
    Moreno J, Lianes P, Cardenal F, et al. Phase II study of docetaxel and cisplatin in first line treatment of disseminated small cell lung cancer: Preliminary results. Proceedings of ASCO 19, 544a. 2000.
  96. ↵
    Schiller JH, Kim K, Hutson P, et al. Phase II study of topotecan in patients with extensive-stage small-cell carcinoma of the lung: an Eastern Cooperative Oncology Group Trial. J Clin Oncol 1996;14:2345–2352.
    OpenUrlAbstract
  97. ↵
    Sorensen M, Jensen PB, Sehested M, Hirsch FR, Hansen HH. Phase I/II study of topotecan and cisplatin alternating with carboplatin, cisplatin, teniposide and vincristine in previously untreated patients with small-cell lung cancer. Proceedings of ASCO 17, 501a. 1998.
  98. ↵
    Kudoh S, Fujiwara Y, Takada Y, et al. Phase II study of irinotecan combined with cisplatin in patients with previously untreated small-cell lung cancer. West Japan Lung Cancer Group. J Clin Oncol 1998;16:1068–74.
    OpenUrlAbstract
  99. ↵
    Masuda N, Matsui K, Negoro S, et al. Combination of irinotecan and etoposide for treatment of refractory or relapsed small-cell lung cancer. J Clin Oncol 1998;16:3329–3334.
    OpenUrlAbstract
  100. ↵
    Noda K, Nishiwaki Y, Kawahara M, et al. Randomized phase III study of irinotecan (CPT-11) and cisplatin versus etoposide and cisplatin in extensive-disease small cell lung cancer: Japan Clinical Oncology Group Study. Proceedings of ASCO 19, 483a. 2000.
  101. ↵
    Ormrod D, Spencer CM. Topotecan: a review of its efficacy in small cell lung cancer. Drugs 1999;58:533–551.
    OpenUrlCrossRefPubMedWeb of Science
  102. ↵
    Fukuoka M, Masuda N. Clinical studies of irinotecan alone and in combination with cisplatin. Cancer Chemother Pharmacol 1994;34:Suppl., S105–S111.
  103. ↵
    Kubota K, Nishiwaki Y, Sugiura A, et al. A pilot study of cisplatin and etoposide plus concurrent accelerated hyperfractionated thoracic radiotherapy followed by three cycles of irinotecan and cisplatin for the treatment of limited-stage small-cell lung cancer: JCOG 9903-DI. Proceedings of ASCO 20, 345. 2001.
  104. ↵
    Graham MV, Jahanzeb M, Dresler CM, Cooper JD, Emami B, Mortimer JE. Results of a trial with topotecan dose escalation and concurrent thoracic radiation therapy for locally advanced, inoperable nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys 1996;36:1215–1220.
    OpenUrlCrossRefPubMedWeb of Science
  105. ↵
    Takeda K, Negoro S, Kudoh S, et al. Phase I/II study of weekly irinotecan and concurrent radiation therapy for locally advanced non-small cell lung cancer. Br J Cancer 1999;79:1462–1467.
    OpenUrlCrossRefPubMedWeb of Science
  106. ↵
    De Marinis F, Cortesi E, Paoluzzi L, et al. Cisplatin, etoposide and gemcitabine in untreated patients with small cell lung cancer (SCLC): Preliminary results of a multi-institutional phase II trial of GOL. Proceedings of ASCO 19, 525a. 2000.
  107. ↵
    Jensen PB, Sehested M, Langer SW, Hansen HH. Twenty-five years of chemotherapy in small cell lung cancer sends us back to the laboratory. Cancer Treat Rev 1999;25:377–386.
    OpenUrlCrossRefPubMedWeb of Science
PreviousNext
Back to top
View this article with LENS
Vol 18 Issue 6 Table of Contents
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Chemotherapy in small cell lung cancer
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Chemotherapy in small cell lung cancer
K. Østerlind
European Respiratory Journal Dec 2001, 18 (6) 1026-1043; DOI: 10.1183/09031936.01.00266101

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Chemotherapy in small cell lung cancer
K. Østerlind
European Respiratory Journal Dec 2001, 18 (6) 1026-1043; DOI: 10.1183/09031936.01.00266101
Reddit logo Technorati logo Twitter logo Connotea logo Facebook logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Treatment strategies in small cell lung cancer
    • Treatment of limited disease
    • Prophylactic cranial irradiation
    • Extensive disease
    • Elderly patients
    • New chemotherapeutic agents
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Asthma remission: what is it and how can it be achieved?
  • Asthma management in low and middle income countries
  • Calcilytics for the management of asthma
Show more Series

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERJ

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • Podcasts
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN:  0903-1936
Online ISSN: 1399-3003

Copyright © 2023 by the European Respiratory Society