109

Critical Review,s in Oncology/Hematology. 1991; 11: 109-135 0 1991 Elsevier Science Publishers B.V. 1040-8428/91/$3.50

ONCHEM

00006

Small cell carcinoma of the lung: clinical and biologic aspects Jean Viallet ‘NCI-Navy

and Daniel C. Ihde’

Medical Oncology Branch, National Cancer Institute and National Naval Medical Center, and Uniformed Services University @‘the

Health Sciences1 Bethesda, MD. U.S.A. and 2the Departments

of Medicine and Oncology and the Montreal General Hospital Lung Oncology Group,

McGill University,

Montreal,

Quebec, Canada

Contents 1.

Introduction

II.

..............................................................

..................... Current status of therapeutic approaches to SCLC ............................. A. Combination and single-agent chemotherapy. . .. . Clinical features,

III.

B. Intensity c.

pathology,

staging,

of induction

Cyclic alternating

D. Duration

of chemotherapy

F. Strategies

factors

110 112 112

chemotherapy

non-cross

E. Late intensification

109

and prognostic

113

resistant

chemotherapy

and management

with autologous

for evaluation

regimens.

113

of tumor progression

bone marrow

114

transplantation.

114

.

of new drugs

.

115

G. In vitro drug testing.. H. Combined

116

modality

I.

Prophylactic

J.

Adjuvant

therapy

and therapeutic surgical

K. Biological

response

L. Modification

B. Interruption

of autocrine

growth

tional Cancer

toxin on signal transduction

fessor in the Departments versity,

Montreal,

Research

Council

from Stanford

logy at Memorial tional Cancer

of Canada.

University. Institute,

Deputy

Chief (Clinical)

National Uniformed

Naval

fellowships

Cancer

Bethesda, Cancer

is a Scholar

Center

Services University

Pro-

at McGill

Uni-

an M.D.

degree

in Medical

Onco-

NY and the Na-

MD. Since 1975, he has been on the

Institute,

where

of the Navy Medical

Medical

Center,

at the Na-

Assistant

of the Medical

Daniel Ihde received

Sloan-Kettering

staff of the National

and Oncology

He completed

of Montreal,

Oncology

and a Professor of the Health

until recently

Oncology

he was

Branch

at the

of Medicine

at the

Sciences,

also in Bethes-

da. Correspondenre: Branch, Bethesda,

National

Daniel Naval

MD, 20889-5105.

C.

Ihde,

Medical U.S.A.

NCI-Navy Center,

Medical

Building

Oncology

8, Room

5101.

123 123

signal transduction pathways

pathways and growth

................

125

..............

126 126 127

....... .... ......... .. .......

MD. He is currently

Dr. Viallet

stimulated

SCLC.......................................................

....... .

in Medical

of Medicine

Canada.

121 therapeutics

.....................................................................

. ... .

Bethesda,

121

as a target for developmental

3. Effects of cholera

a fellowship

Institute,

stimulation

peptide

Jean Viallet received an M.D. degree from the University He completed

121

............................................ ...................................................

of SCLC

2. Gastrin-releasing

References

120

treatment.

survivors.

............................................................

C. Genetic lesions in

Canada.

119 119

with adjunctive

in long-term

mechanisms

I. Growthfactors

Conclusions

II8

.

in the biology

A. Drug resistance

117

irradiation.

modifiers

of host environment

Recent advances

V.

cranial

irradiation

resection

M ‘Cure’ and morbidity IV.

with chest and multiple-site

........ . ..

.

128

I. Introduction Small cell lung cancer (SCLC) distinguishes itself from the other types of lung carcinomas by a remarkable propensity for early systemic spread and by substantially greater initial sensitivity to chemotherapy and radiation therapy. The former is exemplified by the fact that nearly all patients present with mediastinal or systemic metastases at the time of initial diagnosis. Indeed, even patients that were surgically resected but died within 1 month of surgery were found at autopsy to have a high incidence of residual cancer, most often distant metastases [l]. The predominant influence of systemic cancer on survival in SCLC is confirmed by the trivial ef-

110

fects of surgery and radiotherapy on the life-span of all but a handful of patients. Systemic chemotherapy, on the other hand, has increased median survival by 45fold since its introduction in the 1960s [2], even though the impact of chemotherapy on survival in other cell types of lung cancer is marginal at best [3]. Since it comprises approx. 25% of all lung cancer, SCLC is the sixth most common malignancy and the fourth leading cause of death from cancer in the United States [4]. It is the most common neoplasm in which a fraction of patients can be cured with chemotherapy. Substantially more successful management of this tumor would therefore clearly have a significant impact on overall cancer mortality. In the early 196Os, the Medical Research Council of Great Britain conducted a clinical trial in operable SCLC patients comparing surgical resection to locoregional irradiation [5]. Although radiotherapy was found to be superior to surgery in prolonging median survival, neither modality provided long-term tumor control in this relatively favorable group of patients without obvious distant metastatic disease. In contrast, single agent chemotherapy with cyclophosphamide resulted in a doubling of the median survival in both limited and extensive stage patients when compared to placebo treatment in a prospective randomized clinical trial published in 1969 [6]. Administration of this drug to patients with other lung cancer cell types yielded neither substantial tumor regression nor any improvement in survival. Since then, a large number of individual drugs have been utilized in the management of SCLC patients. It was rapidly established that combination chemotherapy was superior to single agent therapy. By the end of the 197Os, drug treatments for SCLC, with or without the addition of chest radiotherapy, were reproducibly associated with median survivals of 10-16 months in patients with localized and 611 months in those with overtly disseminated cancers. Two-year survival of 10-25s and O--3%for localized and disseminated tumors, respectively, was often reported by the early 1980s. These survival results are quite impressive compared to the previously observed median survival of 334 and 1.5-2 months, respectively, in untreated patients [3, 71. There is little data from the pre-chemotherapy era on 2-year survival of unselected patients with SCLC, but it was certainly a rare occurrence. However, there has been little further improvement in survival results over the succeeding 10 years. Figs. 1 and 2 show that this therapeutic plateau has not resulted from inactivity on the part of clinical investigators, as the numbers of both individual agents and innovative therapeutic strategies which have been evaluated have continued to increase. Rather, the generally disappoint-

ing results of these continuing efforts might suggest that empirical selection of drug combinations and variations in their mode of delivery using presently available cytotoxic agents may not be the ultimate answer to the successful management of this disease. During the same decade, in contrast, our understanding of the fundamental biology of SCLC has rapidly expanded and reached a point where it may serve as a basis for the development of novel treatment approaches. In this review, it will be our purpose to trace the evolution of the therapy of SCLC to its current state, with emphasis on areas which have not been sufficiently evaluated, as well as to provide an overview of SCLC tumor biology in an effort to identify potential targets for experimental therapeutics.

II. Clinical features, pathology, staging, and prognostic factors

In most cases of SCLC, it is the location and size of the primary tumor and the presence and nature of metastatic deposits which dictate the accompanying signs and symptoms. Because SCLC tends to present centrally and to infiltrate the submucosa [8], bronchial obstruction with consequent cough, dyspnea, wheezing, chest discomfort, hemoptysis or post-obstructive pneumonia is common. Two-thirds of patients will have evidence of distant metastases, most often to the liver, bone, bone marrow, or brain, at the time of original diagnosis. The clinician must also be alert to signs and symptoms which may be manifestations of the many paraneoplastic syndromes associated with SCLC and which may be confused with symptomatic metastases [9]. These syndromes are often due to the elaboration and release by the tumor of various humoral products (adrenocorticotrophic factor (ACTH) causing ectopic Cushing’s syndrome or arginine vasopressin (AVP) and atria1 natriuretie factor (ANF) [lo] causing hyponatremia and the syndrome of inappropriate antidiuretic hormone (SIADH) secretion) or to immune reactions to a tumorassociated antigen, especially in neurologic syndromes such as the Eaton-Lambert myasthenic syndrome, in which the presence of circulating antibodies reacting with voltage-gated calcium channels has been documented [I 11. Visual deficits produced by retinal degeneration and sensory neuronopathy are two other syndromes most commonly found in SCLC in which serum antibodies reacting with both tumor and neuronal tissue are observed [9]. A diagnosis of SCLC can be made from tissue obtained through a variety of techniques, either from the primary site or a nodal or distant metastasis. Special

111

caution is required in pathologic interpretation of needle aspiration biopsies, however, as crush artifact can sometimes be mistaken for SCLC. The tumor consists of cells that are 2-3-times the size of mature lymphocytes. Nuclei are darkly staining with a characteristic ‘salt and pepper’ chromatin distribution [12]. Small, relatively infrequent dense-core granules, which are a manifestation of the peptide secretion that is the hallmark of the ‘neuroendocrine’ properties which are observed in most SCLC but are relatively infrequent in other types of lung carcinomas, can be demonstrated on electron microscopy in most tumor specimens which are diagnosed as SCLC by light microscopy. Whether these neurosecretory granules are present, however, is not associated with response to chemotherapy [ 19. Whereas the classic oat-cell subtype has little cytoplasm and indistinct or absent nucleoli, the intermediate subtype is composed of somewhat larger cells with more distinct cytoplasm. The distinction between these two subtypes provides no prognostic information, as patients with each of these pathologic diagnoses present with similar extents of tumor dissemination and experience no differences in response to therapy, duration of response, or survival [1416]. However, 5514% of patients present with a tumor composed of classical small cell elements admixed with larger cells with more abundant cytoplasm and prominent nucleoli. This small cell/large cell subtype was of prognostic significance at two different institutions which documented a lesser likelihood of response and shorter median survival in this group of patients [16, 11. This finding, however, could not be confirmed in patients entering a cooperative group chemotherapy trial [18]. A much smaller frequency (2% in one series) of SCLC patients have elements of squamous or adenocarcinoma in their diagnostic biopsy specimens. Although the natural history of patients with this ‘combined’ subtype is not well characterized, they may present more often with surgically resectable lesions P91. Essentially all investigators currently employ the twostage system first proposed by the Veterans Administration Lung Group. The distinction between the two stages is based upon whether the known tumor can be encompassed within a tolerable radiation field, thus introducing physiologic as well as anatomic factors into assignment of stage [3]. Limited stage is defined as disease confined to the hemithorax of origin and its regional lymph nodes (including mediastinal, contralateral hilar and, in most definitions, ipsilateral or contralateral supraclavicular), whereas extensive stage is defined as disease extending beyond this area. Since in many institutions patients with limited-stage disease will receive chest irradiation in addition to systemic chemotherapy,

the definition of stage influences the primary management of the patient. The extent of the staging process varies with the specific clinical situation. In usual practice, presenting signs and symptoms guide the choice of diagnostic procedures. If an unequivocal site of distant metastases is identified, eliminating the indication for elective thoracic irradiation, no further procedures need be performed unless the information sought will modify clinical management. In the rare patient with a proven diagnosis of SCLC who may still be a candidate for surgical resection, preoperative mediastinoscopy should be performed since documentation of mediastinal node involvement would preclude thoracotomy, and the tumor should be staged by the recently revised, more detailed TNM staging system [20]. After therapy has been administered, sites of prior involvement should be reevaluated to determine whether complete, partial, or no objective response is present. The response category is strongly associated with ultimate survival, with virtually no patients without a complete response living beyond 2 years. In the context of a clinical trial, where a careful characterization of the patient population under study is necessary to assure optimal interpretation of the observed outcomes, more detailed and rigorous staging evaluations are usually justifiably pursued. Ambulatory or performance status and tumor stage at presentation are the most powerful prognostic factors in SCLC. In large groups of patients who receive combination chemotherapy, additional factors which refine prognosis can often be demonstrated. Some directly relate to total tumor burden, such as the favorable effect of tumors which were completely surgically resected in limited-stage disease [21] and the number of organ systems involved by metastases in extensive stage patients [22-251. In patients whose tumors are surgically removed, hilar and especially mediastinal node involvement are associated with considerably worse prognosis [26]. Some are host characteristics; women [22, 25, 271 and younger patients (below 60 or 70 years of age) [22, 25,281 have a more favorable outcome in some series or patient subgroups. Finally, abnormal serum biochemical values (elevated liver function tests or low albumin or sodium determinations) provide prognostic information independent of documented tumor extent in many reports [23, 25, 28, 291. Abnormalities in serum biochemical tests are strongly correlated with the number of distant metastatic tumor sites in patients who have undergone numerous systematic staging procedures, and both are likely to be surrogates for overall tumor burden [23, 241. As in all other cancers, SCLC patients who have developed tumor progression after receiving chemotherapy have a

112

markedly inferior prognosis, with median survival of only 24 months [3]. Most, if not all, treatment programs for SCLC are adopted only after documentation of their efficacy in a clinical trial is published in the medical literature. Therefore, it is of great interest to know whether there are any marked differences in prognosis between patients who are entered on clinical trials and those who are not. One report which investigated this issue found little difference in outcome in limited-stage patients whether or not they were placed on institutional clinical trials, but much shorter survival in extensive-stage patients who were not placed on trials because of poor performance status or medical problems unrelated to their cancer [30]. This result is not unexpected, since most limited-stage patients are fully ambulatory, but emphasizes that outcome similar to that reported in clinical trials should not necessarily be expected in more acutely or chronically ill patients. III. Current status of therapeutic approaches to SCLC

The current status of SCLC therapy has recently been extensively and comprehensively reviewed [3, 31, 321. Only the salient points will be summarized here. The ability of combination chemotherapy to substantially improve the clinical outcome of SCLC patients was documented relatively quickly after its introduction, certainly by the end of the 1970s. Since then, clinical evaluation of multiple combinations of drugs, selected both empirically and rationally, and variations in duration and intensity of therapy, including autologous bone marrow support, have had only very modest additional impact on survival. This suggests that future investigations might better focus on the biological mechanisms underlying drug resistance or on alternative forms of therapy diverging from the classical cytotoxic chemotherapy paradigm. III-A. Combination and single-agent chemotherapy

When combination chemotherapy programs were first introduced into the management of SCLC, they appeared to yield much higher response rates, especially complete response rates, than had been observed with single-agent treatment. However, there are relatively few prospective randomized trials which specifically confirm this point. Two- [33] or three-drug [34] regimens containing cyclophosphamide were compared to cyclophosphamide alone, with improved response rates and survival found using the combination programs. Another trial compared concurrent administration of four drugs with the sequential administration of the same

agents and found a superior response rate with the use of the combination [35]. All these trials were published before 1980. They are confirmed by a recent British study which randomized 151 patients to receive combination chemotherapy (with chest irradiation in limited disease) or single-agent cyclophosphamide or palliative radiotherapy whenever indicated. Median survival was doubled from 4 to 8 months and l-year survival increased from 8% to 30% with the combination regimen [36]. The optimal number of drugs in a combination program is not established, but two to four are most commonly given. Since most agents used in the treatment of SCLC are myelosuppressive, each addition to a combination usually requires compromising the dose of all the agents, probably accounting for the fact that little or no benefit has been documented from administering more than three drugs simultaneously [3]. It has proven quite difficult to establish the superiority of any specific chemotherapeutic regimen in the treatment of SCLC. For example, etoposide (VP- 16) as a single agent has one of the highest reported response rates against this tumor, but adding this drug to an effective combination program containing other drugs has generally not led to improved outcome in randomized trials [37-391. In one study in which etoposide was substituted for vincristine in the cyclophosphamide/doxorubicin (Adriamycin@)/vincristine (CAV) combination, survival was significantly improved with etoposide, but only in extensive- stage patients [40]. Etoposide when given as a single agent is more effective on a 5-day than on a lday schedule [41], but when etoposide was added to cyclophosphamide and doxorubicin on either a S-day or 1-day schedule, no differences in outcome could be demonstrated [42], probably because the considerable efficacy of the latter two drugs greatly diluted any therapeutic impact which the addition of etoposide in a superior schedule could provide. A two-drug regimen frequently utilized in SCLC after the mid-1980s is cisplatin and etoposide (PE), which is therapeutically synergistic in murine tumors and extremely effective against testicular cancer [3]. It was first shown to have activity in patients who developed progressive tumor following CAV treatment [43] and then found to yield survival results similar to other regimens when used alone as the only form of chemotherapy [44]. In a randomized study PE produced significantly better survival than ifosfamide/etoposide [45]. In another randomized trial in extensive-stage patients, relatively low doses of PE were equally as effective as 67% higher doses, while producing very modest myelosuppression [46], suggesting that this combination has a very favorable therapeutic index compared to most other combination programs which are administered in SCLC.

113

Thus,

numerous

mens currently

combination

chemotherapy

offer approximately

comparable

regiefficacy

in SCLC patients. The two-drug PE combination appears to be as effective as three- and four-drug programs utilizing

various

combinations

doxorubicin, etoposide, cristine, methotrexate,

of cyclophosphamide,

cisplatin and its analogs, vinand nitrosoureas, the agents

therapy

requiring

prolonged

led to any obvious tions utilized phosphamide,

doxorubicin,

and etoposide

binations,

single agent in a randomized

hematologic

once the three-drug

program

easy to add a third drug toxicity

to this program,

has been shown

to have

greater efficacy. At present, the choice of a specific combination regimen can be appropriately tailored to take into account

the expected

tolerance

of an individual

pa-

tient to drug-specific toxicities other than myelosuppression, as well as cost and ease of administration. Although combination chemotherapy is the treatment of choice for SCLC at present, some recent studies, most of them performed in Europe, have administered a single-agent podophyllotoxin derivative, either etoposide or teniposide, as sole therapy for previously untreated, generally elderly patients [47.48]. Response rates of 8& 90% were observed, with median survival of approx. 9 months and very modest toxicity. Whether this result is equivalent to what would have been obtained with combination chemotherapy is unknown, but such therapy may appropriately be considered for elderly or poor performance status patients, particularly with extensivestage disease. Prolonged oral administration of oral etoposide, which can maintain cytotoxic serum concentrations of the drug for protracted periods, has also been investigated in previously untreated patients [49]. III-B.

Intensity

of induction

chemotherapy

With the exception of some programs of PE, most current chemotherapy regimens are designed to produce moderately severe myelosuppression, their dose-limiting toxicity (nadir white blood cell count of lOOO-2000/~1 in the majority of patients). Some early randomized studies utilizing cyclophosphamide-based regimens supported the concept that higher doses of drugs were associated with a modest but significant improvement in survival [50, 511. Extremely high doses of single-agent cyclophosphamide (200 mg/kg) combined with chest irradiation have also yielded an apparently increased fraction of complete responses and survival similar to patients given more usual chemotherapy in limited disease [52]. However, improved survival with higher initial drug doses has not been demonstrated in randomized studies employing the CAV program [53, 541 or PE [46]. Tnuncontrolledstudies,evenhigherinitialdosesofchemo-

not

Combina-

[55-571 or cy-

clophosphamide, cisplatin, and etoposide [58]. The failure of etoposide-containing high-dose programs to yield markedly superior results is consistent with the lack of improvement doubling or tripling

it should be relatively

have

in survival.

in these trials have been based on cyclo-

most commonly utilized in SCLC [3]. Since it is also somewhat less myelosuppressive than most other comwith substantial

hospitalization

improvement

in response rates or survival with of the dose of etoposide given as a

rent SCLC [59]. In summary, vincing more

evidence than

that

moderately

trial in patients presently

treatment

with recur-

there is no con-

designed

to produce

severe myelosuppression

at the

time of induction yields better results, and such therapy should be administered only in the setting of a clinical trial.

III-C.

Cyclic

chemotherapy

alternating

non-cross

resistant

regimens

Goldie et al. have proposed an elegant mathematical model of the emergence of chemotherapy-resistant clones in malignancies [60] which predicts that superior clinical efficacy would be expected with the earliest feasible administration of as many active drugs as possible. Because of the partially overlapping toxicities of most regimens, this goal can be best achieved with the least compromise in doses of individual agents by cyclic alteration of non-cross resistant drug combinations. However, if we accept as a reasonable definition of non-cross resistance the ability to induce complete remissions after tumor progression while receiving primary treatment, no current chemotherapy programs for SCLC can be considered non-cross resistant. It is, therefore, not surprising that most randomized clinical trials comparing this strategy to the standard approach of treating with a single regimen until therapy is completed have generally failed to yield superior survival results [3], although in some studies duration of initial remission was prolonged [61]. A more recent trial conducted by the National Cancer Institute of Canada randomized extensive-stage patients to receive either the CAV combination for six cylces or CAV alternating with PE for the same duration of treatment [62]. Patients given the alternating regimen experienced a 6-week prolongation in median survival which was statistically significant. However, it is not clear whether this modest improvement was due to the alternating strategy or to an inherent superiority of PE, especially since in another randomized trial conducted in limited-stage patients the addition of only two cycles of PE given after completion of six cycles of CAV led

114

to a 7-month improvement in median survival compared to no further therapy [63]. Two more recent, larger studies comparing CAV to PE to alternation of the two programs should help to resolve this issue, although they include relatively few patients with limited disease. Preliminary results indicate no substantial survival differences [64, 651. At present, the use of noncross resistant chemotherapy regimens is an available option in the primary management of SCLC. Its bestestablished virtue at this time is not in offering clearly superior survival but rather in permitting reduction in toxicities dependent upon the cumulative dose of individual agents, such as cardiotoxicity of doxorubicin or neurotoxicity of vincristine and cisplatin. III-D. Duration of chemotherapy tumor progression

and management

of

Early chemotherapeutic programs for SCLC were frequently administered for up to 2 years in the few patients in whom tumor progression had not occurred by that time. However, it has now become clear from both uncontrolled data [66] and most prospective randomized trials [67-69] that prolonged maintenance chemotherapy does not extend survival, although the duration of initial remission may be prolonged [70]. One large trial of 610 patients was able to assess the survival benefit of administering chemotherapy at relapse in a randomized fashion [67]. Survival was prolonged with such ‘salvage’ treatment in patients receiving only four cycles of initial therapy, but not in those given eight cycles. At present, induction chemotherapy lasting 12 to 18 weeks would appear to be optimal. One method to increase delivery of drugs, while still reducing duration of therapy, is weekly administration of myelosuppressive and non-myelosuppressive drugs on an alternating basis. This approach appeared promising in a pilot study [71]. In addition to worsening the morbidity of treatment, maintenance chemotherapy may also reduce the likelihood of palliation from drug regimens given for relapsing tumor. PE chemotherapy produced much lower response rates in one study conducted in patients with recurrent SCLC who had a median ‘drug-free’ interval of less than 1 month since their most recent prior treatment than was observed in other trials of PE in which, due to the policy of avoiding prolonged maintenance therapy, the median time since previous therapy was 3 to 5 months [72]. Much higher response rates in previously treated patients who had not received chemotherapy for at least 10 to 12 weeks were also noted in Phase II studies of teniposide [73] and daily oral etoposide [74].

III-E. Late intensiJication with autologous bone marrow transplantation

Most patients with SCLC who achieve remission with currently available therapies will relapse and die from their disease, indicating the persistence of occult tumor deposits even in clinically complete tumor regression. Given this context, various investigators have proposed the use of high-intensity therapy after induction with conventional doses of agents to take advantage of treating patients at a time when tumor bulk is at its lowest and the physiological status of the patient optimized. At least nine trials have explored this strategy with a variety of high-dose chemotherapy regimens, usually with autologous bone marrow support and sometimes with the addition of regional or total body irradiation [75]. Most included both limited-stage and extensive-stage patients, and some included drugs which had already been administered at more conventional doses during the induction phase in the late intensification regimen. Only 18-38s of patients beginning therapy with the intent of eventually administering late intensification actually received the high-dose treatment. Although some partial responses were converted to complete responses during the late intensification phase, these responses tended to be of short duration. Importantly, relapses at the primary site in the absence of chest irradiation were prevalent even in patients who were in complete remission prior to intensification, suggesting that the late high-dose chemotherapy by itself had very modest ability to eradicate occult tumor. Overall survival in these uncontrolled trials was not obviously different from that of patients receiving conventional therapy [75]. In the only study utilizing a prospectively randomized design, remission duration (though not survival) was prolonged with late intensification, but survival of all patients beginning induction chemotherapy was similar to most reports of treatment without intensification [76]. Late intensification should be administered only in the setting of a clinical trial. Given the very limited benefits of this approach thus far, future studies might best focus on the limited-stage patient who has achieved a complete remission withconventionalchemotherapy and include thoracic irradiation, probably after intensification, to better control the primary site [77]. Care should be taken to utilize autologous marrow support only if necessary, because of the potential risk of reinfusing marrow contaminated with viable tumor cells 1781. Because of unavoidable selection biases in patient entry onto such toxic therapeutic programs, the value of dose-intense approaches in initially responding patients can most likely become established only by randomized clinical studies.

115 2All antibody G-CSF Am&de

Iproplatin RA233 Idambicin GM-CSF Lonidamine y-interferon DFMO Aclarubicin Propionobacterium 6-M-urine Zinostatin BtitlWle Mitoguazone zII a-interferon Mitoxantrone VinbIastine Aziridinylbenzcquinone Pmcart&ne Maytansine DON Chlorozotocin Amsacrine Vindesiie PALA 5-Fu ACNU DianhydrogaIactilol Warfarin TriaziIlate Prednimustine BCG MER High-dose methouexate Thymosin Cisolatin

Dibromodulcitol

Sueptozotocin Mitomycin C Procarbazine

63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90

Fig. 1, Perspective

on the evaluation

of single agents in the treatment

of SCLC. Data were abstracted

from the reviews of Cohen and Perevodchikova

[291] and that of Joss et al. [292] as well as from selected primary references [85-87, 148-l 50, 165, 169, 170, 173, 174, 293-2951. have well documented activity as single agents.

III-F.

Strategies for evaluation

of new drugs

At least eleven drugs have been shown to have measurable activity when given as single agents to patients with SCLC (Fig. 1). At a time when therapeutic results have been almost static for the last decade, it is disappointing that most if not all efficacious drugs identified during this period, such as carboplatin and epirubicin, are structural analogs of agents which were already known to be active at the time the newer drugs were introduced.

Drugs in boldface

Patients who progress in the face of or shortly after receiving induction or subsequent chemotherapy programs seldom experience a meaningful response to additional treatment. In this setting, even drugs quite useful as primary therapy, such as etoposide [79] and cisplatin [80] may have minimal or no detectable activity. Because of this, there is justifiable concern that the investigation of new drugs in previously heavily pretreated patients, as was customary until relatively recently, may lead to the inappropriate classification of truly active agents as being inactive [8 11.

116

Since the ultimate value of a new drug can only be determined in the clinic, novel strategies have been proposed to optimize the evaluation of Phase II drugs. These include the study of relapsed patients while lowering the cut-off point for a Phase II study response rate that is considered promising to 10% [82], enrolling only patients who relapse after a certain interval free of treatment [83], and testing novel agents as initial therapy in newly diagnosed patients [84]. The latter strategy is probably the most efficient in terms of estimating the true activity of a novel compound with the fewest patients. However, some uncontrolled studies employing this policy have reported that it may be deleterious to the ultimate survival of the enrolled patients, after observing a compromised response rate to standard therapy following initial exposure to an inactive investigational agent [85]. Recent Phase II trials using more stringent entry criteria have evaluated new drugs, including the active agent epirubicin [86] and the inactive agent amonafide [87], with overall survival comparable to that of patients treated conventionally. Most convincingly, a randomized trial of the Eastern Cooperative Oncology Group (ECOG) compared the ultimately inactive agent menogaril to CAV combination chemotherapy in good performance status extensive-stage patients in no need of immediate palliation of tumorrelated symptoms with strict rules requiring cross-over from the investigational agent to a standard combination regimen whenever clinically indicated, without observing any compromise in survival [88]. Initial therapy of extensive-stage SCLC patients with an investigational single agent appears to be a reasonable approach if there is reason to believe that the agent to be tested may be active and that the patient population is selected in such a way that an initial lack of response is not likely to be associated with immediately life-threatening tumor progression. The detailed criteria of Ettinger et al. [89] and Cullen et al. [84] provide generally adequate guidelines for this trial design. The recent suggestion by the National Cancer Institute of Canada Clinical Trials Group that a trial should be terminated if the initial experience rules out a minimal response rate of 30% is attractive [87]. It would limit the number of patients exposed to ineffective therapy and focus the drug development process on the most acitive agents. At present, such a policy appears to be the most promising way to identify novel active drugs for SCLC, which are sorely needed. III-G. In vitro drug testing

If new, more effective drugs are not immediately forthcoming, it may be possible to optimize an individu-

al patient’s therapy by selecting the currently available agent(s) to which his tumor is the most sensitive. This concept was recently tested by our group in a prospective study of extensive-stage SCLC in which a concerted effort was undertaken to establish a cell line from clinical tumor specimens obtained at the time of initial diagnostic and staging procedures [go]. Because of unavoidable delay in obtaining sufficient cultured tumor cells to perform in vitro sensitivity testing with seven drugs known to be active in SCLC, the 80 patients entered in the study were initially treated with PE chemotherapy for 12 weeks. Patients achieving partial remission or experiencing no response, as well as those patients suffering a relapse after an initial complete remission, were treated with a three-drug combination based on in vitro drug testing of their own tumor, if that information was available, or alternatively with the standard CAV combination. Despite being able to obtain tumor-containing specimens for cell culture in 75% of patients, cell lines could be established and drugs tested in only 33%. Ultimately, 20% of all patients received a regimen based on in vitro sensitivity testing. Of interest, in vitro drug sensitivity was significantly correlated with clinical response to both primary and second-line therapy. However, because SCLC cultures tended to be either generally sensitive or generally resistant to most drugs evaluated, the selection of a second-line regimen with in vitro drug testing did not translate into an obvious therapeutic advantage; 4/ 16 (25%) of patients receiving an ‘in vitro best regimen’ achieved further remission, whereas 3/43 (7%) treated with CAV did so. This modest difference was not statistically significant. In vitro drug testing as performed in this study was of only minimal clinical value. The labor-intensive technology employed and the lack of marked heterogeneity in sensitivity among cell lines to the standard drugs tested currently limit in vitro sensitivity testing to select drugs for individual SCLC patients to investigational studies. However, the correlations observed between in vitro and clinical drug sensitivity, which have now been maintained for a median of 27 months in cell culture [91], suggest that these or similar cell lines taken directly from SCLC patients might be useful for studying mechanisms of drug resistance or in screening for new chemotherapeutic agents. The National Cancer Institute, through its Lung Cancer Drug Discovery Project and other drug screening programs, has reorganized its strategy for the identification of novel active agents [92]. It is hoped that a panel of human lung cancer cell lines of diverse origins, used in an in vitro screening process, will identify compounds with tumor type-specific activity which could more frequently be documented to possess clinical efficacy

117 Growthfacta antagonists In vitro drug testing Hypeaftxtiarated cheat Won “Adjuvant” surgical reacction Bone marrow stimulation with CSFs High intensity induction Hypxalimentation Mainturancecllem~ Autologous bone marrow transplant AnticoagoMon Immimotkaapy Cyclic alternating chemotherapy Prophylactic cranial irradiation Comb&on chemotherapy Chemomdiotheraov for LD Single agent chemotheqy

FvimarY ldialioll themov .SurRery

63 64 65 66 61 68 69 70 71 72 73 74 75 76 II

Fig. 2. Perspective introduction

on the evolution

of the indicated

of treatment

modalities

modalities

was chosen

78 79 80 81 82 83 84 85 86 87 88 89 90

for SCLC.

from among

The publication

available

colony-stimulating

than agents previously identified through initially screening against highly sensitive murine leukemias. III-H. Combined modality therapy with chest and multiple-site irradiation

The refinement of combined modality treatment with chemotherapy and chest irradiation is the area of investigation which appears to show the greatest promise in terms of improving the outcome of limited-stage SCLC patients in the near future. Since radiotherapy can induce regression of thoracic tumor in up to 90% of cases and was the standard treatment for SCLC at the time combination chemotherapy began to be administered for this .disease, initially many programs included chest irradiation as a component of treatment. A review of published literature in 1981 revealed, perhaps not surprisingly, that chest radiotherapy seemed to add little to therapeutic results that could be achieved with chemotherapy alone in patients with extensive stage disease. In limited disease, although there was little difference in reported median survival whether or not irradiation was given in conjunction with chemotherapy, 2-year survival appeared superior with radiotherapy [93]. However, biases in patient selection could have accounted for this result in this uncontrolled data. In the past decade, long-term results of at least seven large randomized trials comparing chemotherapy given with or without thoracic irradiation in limited-stage SCLC have become available and help to clarify this issue. The majority of these studies found that the addition of chest radiotherapy to systemic chemotherapy leads to an improvement in complete response rate, control of the primary tumor, and disease-free survival and overall survival [94-971, although several [98-1001 did

references.

date of the paper Abbreviations

felt to be most representative

used are: LD, limited

disease;

of the

and CSF’s,

factors.

not. Differences in the temporal relationship between radiotherapy and chemotherapy among the trials may account for variations in the effectiveness of irradiation. In general, studies in which chemotherapy and radiotherapy were given either concurrently or in an altemating fashion without any delay in the scheduled administration of chemotherapy were much more likely to document survival benefit from radiation than studies in which the two modalities were given sequentially, with a planned interruption in chemotherapy in order to deliver the irradiation [3]. The survival advantage with radiotherapy in positive studies has been relatively modest, with l- to 4-month improvement in median and 717% absolute increase in 2-year survival. However, advantages of this combined modality approach have been somewhat compromised by the associated increase in toxicities such as myelosuppression, esophagitis, weight loss [3], and in particular severe pulmonary toxicity which in one trial accounted for fatalities in complete remission of 10% of patients [loll. Many trials used chemotherapy regimens which included agents with a potential for lung damage. Unfortunately, the apparently more efficacious concurrent combined modality programs are usually more toxic than sequential ones. Although only a few controlled [96] and uncontrolled [102] studies have been performed, rapidly alternating programs of chemoradiotherapy may have reduced pulmonary toxicity while preserving the probable survival advantage of concurrent combined modality approaches. The basic radiobiological observation that SCLC cell lines had a reduced ability to recover from low doses of radiation when compared to normal cells has led to a novel approach using twice-daily administration of radiation therapy [103]. In pilot trials in combination with

118

PE chemotherapy, this approach appeared both highly effective, with median survival in excess of 2 years, and better tolerated than previous regimens [104, 1051.These experiences from single institutions were recently extended through the ECOG, which treated 40 patients in this fashion; a 95% overall response rate was observed, with 3 1 complete remissions [ 1061.There were four toxic deaths, two of them pulmonary, but 2-year actuarial survival was 36%. A randomized trial designed to assess the contribution of twice-daily radiation therapy compared to a standard once-daily concurrent combined modality approach with PE is currently underway, since it is possible that PE chemotherapy itself is especially well suited for concurrent administration with chest irradiation, being both less toxic and perhaps more effective [ 1071, and accounts for the apparently better results of the twice-daily programs. Some combined modality programs thus appear to produce improved survival in limited-stage SCLC, but these are technically complex in their delivery and require close cooperation between radiation and medical oncologists. Since not all combined modality treatments increase survival, but essentially all increase toxicity, chest irradiation is not mandatory in all patients, especially those with poor pulmonary function or performance status. For other patients not participating in clinical trials, concurrent radiotherapy and PE chemotherapy are at present most often recommended. Irradiation to extrathoracic sites other than brain in conjunction with chemotherapy for SCLC appears to yield no clinical benefits, either prophylactically in limited disease [108] or to the primary tumor and clinically involved extrathoracic sites in extensive disease [ 1091. Thoracic radiotherapy alone given with chemotherapy to extensive-disease patients can reduce the frequency of tumor progression in the chest but appears to have no effect on survival [ 1lo]. III-I. Prophylactic and therapeutic cranial irradiation

Patients receiving chemotherapy for SCLC have a tendency, which progressively increases over time, to develop central nervous system (CNS) metastases, especially in the brain. In one series, the actuarial likelihood of CNS metastases in the absence of specific therapy was approx. 80% at 2 years [l 111. Because these findings were similar to what had previously been observed in childhood acute lymphoblastic leukemia, prophylactic cranial irradiation (PCI) was also proposed as a potentially useful maneuver for SCLC. This topic has recently been comprehensively reviewed [ 1121. A combined analysis of the nine published randomized studies assessing the impact of PC1 in

SCLC shows a definite ability of this intervention to reduce the incidence of clinically detected brain metastases from 23% to 6%, without, however, affecting survival. Retrospective analysis indicates that the benefits of PC1 are principally restricted to patients who attain a complete remission of their cancer [113], a not unexpected result since if extracranial tumor persists after PCI, it would still have the potential to continue metastasizing to the brain. Although little data addresses this point, a survival benefit does not appear to exist for PC1 therapy even if analysis is restricted to patients in complete remission [113, 1141. The obvious value of reducing the development of brain metastases must be balanced, however, against the possibility of late neurological side-effects associated with the administration of PCI. Combining data from eight studies of long-term survivors of SCLC, most of whom (102/123) had received PCI, showed a 45% incidence of symptomatic organic brain syndrome, which was most commonly manifested by memory loss, gait abnormalities, and coordination defects [112]. Most of these patients also had received long-term chemotherapy regimens which included potentially neurotoxic drugs such as methotrexate and nitrosureas. We [115] recently updated our observations in long-term survivors 6-13 years after therapy for SCLC and found that the high incidence of neuropsychological sequelae observed in an earlier evaluation performed 4 years previously had often continued to progress, albeit not dramatically, in some patients. It should be emphasized, however, that most neurologic morbidity observed in long-term survivors does not compromise patients’ ability to care for themselves [116, 1171, and whether all of the neurologic morbidity observed in these patients is directly related to PC1 is by no means established. Whether PC1 in the context of short-term, cisplatinbased chemotherapy is associated with the same longterm sequelae is unknown at present. In the meantime, it seems prudent to limit the administration of PC1 to patients who achieve complete remission of their tumor and consent to receive it after becoming thoroughly aware of benefits and potential risks. Treating with low doses of irradiation per fraction only after all other therapy is completed may be associated with less neurologic toxicity [ 118-J. Most SCLC patients who develop brain metastases during or after chemotherapy do so in the setting of progressive cancer in multiple sites. Therapeutic brain irradiation in the majority of cases is able to relieve or reduce neurologic symptoms for the remainder of the patients’ usually short life expectancy. However, symptomatic recurrent metastases after irradiation are a common problem in patients who survive for several months

119

or longer [119, 1201. Therefore, attempts to achieve more durable control of intracranial tumor should be considered in patients who have brain metastases as their sole site of extensive disease at the time of diagnosis [121, 1221 or those who relapse from complete response solely in the brain. It has recently become apparent that systemic chemotherapy without cranial irradiation may produce regression of brain metastases which are present at the time of diagnosis [123, 1241, presumably because the bloodbrain barrier to penetration of chemotherapeutic agents is disrupted in areas of gross metastases. What effect this knowledge will have on policies for treatment of brain metastases remains to be seen. III-J. Adjuvant surgical resection

The early experience with surgical resection in the treatment of SCLC was disappointing. The Medical Research Council trial, which randomized potentially resectable patients by the standards of the day, which included much less extensive staging procedures, to undergo surgery or radiation therapy had no long-term survivors in the surgery arm [5]. These and other [125] observations led to the abandonment of surgery in the primary management of this disease. However, more recent reports have challenged this attitude by demonstrating favorable long-term survival in patients with localized presentations (Tl-T2, NO or Nl) treated with initial surgery with or without additional chemotherapy and radiotherapy [21, 26, 126, 1271. Whether this relatively good prognosis is related to the therapeutic effects of surgical resection, the smaller overall tumor burden, or the inherent less aggressive biological behavior of what some have suggested to be more differentiated neuroendocrine lung carcinomas [ 1281 remains unclear. However, it is clear that concern about discovering an SCLC histology should not motivate heroic efforts at obtaining a preoperative diagnosis in the case of a patient presenting with a peripheral lung lesion and no contraindications to surgery. Should such a lesion prove to be SCLC, it should be resected if otherwise appropriate provided mediastinal nodes are not involved, and the patient should then receive six cycles of adjuvant chemotherapy, possibly with chest irradiation. If a diagnosis of SCLC is known preoperatively in a patient who appears to be a good surgical candidate, thoracotomy should be considered only after a negative mediastinoscopy or mediastinotomy. A more controversial area is whether surgery should be employed for resection of residual disease after a maximal response is achieved with chemotherapy. The rationale for this approach is based on the observation

that the site of first relapse in patients achieving a complete remission is often the original site of disease in the chest. It is reasoned that surgical resection of the initially involved lung might decrease the incidence of recurrence and improve survival. Many uncontrolled series have been published [ 129-1331, some of which reported a decreased rate of thoracic recurrence in patients undergoing surgical resection [ 129, 132, 1341. Resected patients experience overall survival much better than reported for limited-stage patients in general; however, this would be expected since they have tumor of relatively small extent which can be completely surgically extirpated. The impact of surgery on these favorable outcomes is currently the object of a prospective randomized study conducted by the Lung Cancer Study Group. Uncontrolled data suggest the major benefit of post-chemotherapy surgery is confined to patients without mediastinal lymph node metastases at diagnosis [ 1351. III-K. Biologic response modifiers

Various immune deficits have been observed in patients with SCLC. Initial reports focused on depression of total lymphocytes and T-cell levels [136], as well as a decreased response to DNCB skin testing [137]. Impaired response to delayed hypersensitivity skin testing has been associated with impaired survival in patients receiving chemotherapy, especially those with otherwise favorable prognostic factors [ 1381.More recently, significantly decreased numbers of T-helper and T-suppressor subsets of circulating lymphocytes [ 1391, as well as a decreased proliferative response of peripheral blood lymphocytes to phytohemaglutinin and interleukin-2, have been described [140]. Although some of these findings are likely to be secondary to the generalized debility associated with an advanced malignancy, the coexistence of pulmonary [ 14 1, 1421and extrapulmonary [ 1431small cell carcinomas in cases of the acquired immune deficiency syndrome raises the possibility that these immune deficits might participate in the genesis of SCLC. However, it should be noted that other authors have found an increase in activated T cells in the peripheral blood of SCLC patients [ 1441,and that in two immunotherapy trials, lack of response to a panel of skin test antigens was of no prognostic import [ 145, 1461. The possible contribution of an immune deficit to the pathogenesis or progressive nature of SCLC led to a number of trials using non-specific immunotherapy in this disease. Initial non-randomized studies were inconclusive [ 1471.Since then a number of randomized studies evaluating the addition of different forms of immunotherapy to various combination chemotherapy regimens, often combined with thoracic irradiation, have

120

been reported. Propionobacterium granulosum strain KP-45 [148], bacillus Calmette-Guerin (BCG) [149], and the methanol extractable residue of BCG (MER) in various schedules [145, 146, 1501 have been studied. These trials have included a total of 870 patients; all have shown no significant difference between standard treatment and immunochemotherapy in terms of response rate, duration of response, or survival. However, significantly increased morbidity was observed with the use of MER. In addition, an early randomized trial suggesting survival benefit (but no effect on the response rate) with the addition of thymosin fraction V to chemotherapy [ 15 l] could not be confirmed [152]. These data indicate that non-specific stimulation of the depressed immune function observed in SCLC does not alter the course of this disease or its response to therapy. In an effort to identify targets for specific immunotherapy, various groups of investigators have generated a plethora of murine monoclonal antibodies after immunization of animals with SCLC tumor preparations. The antigenic specificity of most of these reagents remains undefined, but there appears to be overlap among several of them. To try to bring some order to this field, an international workshop was held in 1987 and led to a grouping of most antibodies within five clusters, each of which detected related antigenic characteristics [ 1531. It was concluded that no antibody showed absolute specificity for SCLC, although some appeared to be relatively specific for neuroendocrine tissue. The ultimate usefulness of these reagents as tools in diagnosis, immunodetection, or therapy remains undefined. In vitro studies suggest that some of these monoclonal antibodies might be useful for the eradication of occult cancer cells from bone marrow in preparation for autologous transplantation [154156] or as potentiators of lymphokine-activated killer cell activity against SCLC [ 1571. Perhaps of more interst is the reduced expression of specific antigens on the surface of SCLC cells, namely the class I major histocompatibility antigens. Since these molecules play a crucial role in immune recognition, their lack of expression may be relevant to the escape of SCLC from putative immune surveillance and contribute to its metastatic potential. Doyle et al. [158] found that SCLC cell lines, in contrast to other lung cancer lines, are deficient in the expression of HLA-A, B, and C antigens as well as of µglobulin, but that treatment with leukocyte-A interferon as well as y-interferon could induce the expression of these antigens. However, in a different panel of SCLC cell lines, Ball et al. [159] were able to detect expression of class I and II major histocompatibility antigens but did note increased expression after treatment with y-interferon. In addition, they and others [160] observed decreased pro-

liferation of SCLC in response to y-interferon. More recently, it was shown that y-interferon-induced modulation of class I major histocompatibility antigen expression in SCLC is associated with increased lysis by cytolytic T lymphocytes [161] but decreased sensitivity to natural killer cells in vitro [162, 1631. The various interferons have not been extensively evaluated in the treatment of SCLC. However, thus far no significant tumor regression has been observed with either cr-interferon [ 1641 or y-interferon [165] in small Phase II trials. However, as has been suggested [166], interferons theoretically might be more useful in maintaining response in patients experiencing a complete remission on chemotherapy. Preliminary results of a randomized trial in Finland testing the efficacy of low-dose a-interferon in SCLC patients in complete remission demonstrate modestly but insignificantly better survival with cx-interferon compared to continued chemotherapy or no further treatment [167]. An additional randomized trial evaluating this treatment is underway. III-L. ModiJication of host environment with adjunctive treatment The frequent weight loss observed at diagnosis in SCLC patients is a factor of negative prognostic significance [28]. Because of this, attempts to improve prognosis by administering parenteral hyperalimentation during chemotherapy have been evaluated in randomized trials. These have failed to show improvement in response rates, survival, or chemotherapy-induced morbidity [168], despite improvements in body weight and reversal of anergy to skin test antigens in another study [169]. Another strategy which has generated interest since the initial description of its antimetastatic effects in some animal tumors has been the use of anticoagulation as an adjunct to chemotherapy. An initial trial conducted by the Veterans Administration Lung Group randomized 50 patients with limited, extensive, and undefined disease extent to receive chemotherapy and thoracic irradiation with or without warfarin anticoagulation. The 25 patients randomized to standard treatment experienced a median survival of 24 weeks, whereas those receiving adjunctive anticoagulation had a median survival of 50 weeks. There were no differences in response rates, which were low (28 and 32% overall) in both arms [170, 1711. The Cancer and Leukemia Group B (CALGB) explored this issue within a larger randomized trial in which 86 extensive-stage patients received combination chemotherapy alone and 103 received the same chemotherapy with warfarin. Despite a statistically superior overall response rate in the warfarin arm,

121

there was no significant difference in survival [172]. The results of a similar CALGB trial in limited-stage patients are awaited. In the meantime, warfarin should not be part of therapeutic interventions in SCLC except in clinical trials. In addition, the putative antiplatelet agent mopidamol (RA-233) was not found to improve the results of combination chemotherapy in another randomized study [173]. The availability of recombinant human granulocyte/ macrophage colony-stimulating factor (GM-CSF) has raised the possibility of reducing chemotherapy-induced neutropenia, the cause of much of the iatrogenic morbidity associated with treatment of SCLC. In vitro studies have suggested that SCLC growth could be inhibited by this agent [160]. Preliminary uncontrolled trials supported the notion that GM-CSF can be administered safely to SCLC patients, and that it does in fact ameliorate myelosuppression [174, 1751. An interim analysis of a large randomized study designed to test the effects of granulocyte-CSF (G-CSF) on myelosuppression induced by the combination of cyclophosphamide, doxorubicin and etoposide has been reported. G-CSF administration was associated with a significant reduction of the frequency and severity of neutropenia. The number of days of hospitalization and antibiotic therapy required for febrile neutropenia were reduced by 4&50$, for the chemotherapy cycles including G-CSF, compared to cycles during which patients were receiving a placebo [ 1761. The role of this novel approach remains to be defined. Given the treatment-related death rates from current SCLC therapy of 0-4s in limited and 2-8s in extensive disease [3], the potential for improving survival with the addition of CSFs to standard chemotherapy is negligible. Their efficacy in reducing the frequency of fever and documented infection compared to prophylactic antibiotics should be investigated. It is possible that CSFs will permit escalation to a higher level of dose intensity of chemotherapy, which may allow the attainment of better therapeutic results with at least some drugs [177]. However, it must be noted that some current regimens such as PE achieve therapeutic results at least very similar to those of more myelotoxic programs based on combinations of alkylating agents and doxorubicin.

reports from six institutions or groups describing survival of a total of 2000 SCLC patients followed for a minimum of 5 years from the initiation of chemotherapy revealed that 4% of all patients, including 7% with limitedand 1% with extensive-stage disease, were alive at the 5year point [31]. Since the probability of recurrence of the original tumor is 33% after disease-free survival of 24 months, 26% after 30 months, 14% after 36 months, and rare after 5 or 6 years [178-1801, 5-year survival, as is true in other types of lung cancer, is a reasonable benchmark for ‘cure’ of SCLC. This cancer can therefore be eradictated by current therapy in a small fraction of patients. Whether such patients should be considered to be cured of their disease, however, is dependent upon the definition of cure. Patients who are free of SCLC 18 to 24 months after the beginning of therapy have a IO-fold increased risk of dying over the next 5 years compared to the general population [180] and a 6-fold increased risk of dying of non-neoplastic causes [178]. The major causes of this excess mortality are principally smoking-related, as was the original SCLC, and include cardiopulmonary disease and second malignancies, the most common of which is non-small cell carcinoma of the lung [181]. Chemotherapy-related acute myelogenous leukemia is rarely observed [182], but this complication should become even more uncommon since the customary duration of chemotherapy is being shortened and nitrosoureas and procarbazine, which are highly leukemogenic drugs, are being utilized much less frequently in treatment of SCLC. Neuropsychological deficits noted in some long-term survivors have been discussed in Section III-I. Although these second malignancies and chronic toxicities can be devastating in some individuals, they are clearly outweighed by the several-fold prolongation in median survival for all patients receiving current therapy and the small but real prospect of permanent eradication of their cancer for a few. Despite the major advances made in the management of SCLC in the chemotherapy era, however, it is sobering to reflect that the great majority of patients still die of their cancer.

III-M.

IV-A.

‘Cure’ and morbidity

in long-term survivors

Although the improvement in median survival of SCLC patients given combination chemotherapy was obvious almost immediately after its utilization became widespread in the 1970s the question of whether any patients are ultimately cured of their disease began to be answered only within the past few years. A summary of

IV. Recent advances in the biology of SCLC Drug resistance mechanisms

The empirical chemotherapy of SCLC, despite remarkable early successes, appears to have reached a plateau. One possible approach to breach this impasse is to seek an understanding of the mechanisms by which SCLC cells escape the toxic effects of the diverse classes of agents to which they have been exposed. This knowl-

122

edge might then be applied to novel strategies designed to neutralize or bypass these defense mechanisms. The study of drug resistance of cancer cells has attracted much attention in recent years. The earliest data were obtained on the mechanisms of resistance to the antimetabolite methotrexate. Before reaching its major intracellular target, the enzyme dihydrofolate reductase, methotrexate must enter the cell through an energy-dependent specific carrier, after which its intracellular accumulation is facilitated by polyglutamation. Sensitivity to dihydrofolate reductase inhibition depends in part on the continuous consumption of reduced folates by thymidylate synthesis. As a consequence of these complex events, resistance to methotrexate can result from alterations in many different aspects of the drug’s interactions [183]. In SCLC cell lines, Curt et al. [ 1841were able to correlate relative methotrexate resistance with reduced intracellular drug accumulation due to decreased polyglutamation, as well as decreased thymidylate synthetase activity. In addition, unstable methotrexate resistance in cultured SCLC cell lines was occasionally found to be due to elevated levels of dihydrofolate reductase caused by gene amplification associated with the presence of double-minute chromosomes [ 1851. Although these early observations emphasized the ability of neoplastic cells to develop mechanisms to escape the toxic effects of specific chemotherapeutic agents, they could not address the broader question of concomitant resistance to diverse classes of agents so often observed in the clinic. Variants of the ‘multidrug resistance’ phenotype have been attributed to the highlevel expression of a drug efflux pump, the ~170 glycoprotein encoded by the multi-drug resistance gene MDRl[l86-1891, the expression of efficient drug-detoxifying enzymes such as the placental form of glutathione transferase [190], or an enhanced ability to repair DNA damage [183]. A particularly useful resource to address these issues is the large panel of lung cancer cell lines now in existence. Studies with these lines, derived from both previously treated and untreated patients, should allow the eventual correlation of drug sensitivity patterns with cellular mechanisms of drug resistance. Our group has studied a population of 68 consecutive extensive-stage SCLC patients in whom systematic efforts were made to harvest tumor tissues and establish a cell line from each patient [191] and found no correlation between the ability to establish a cell line in the 23 patients in whom that could be accomplished and their clinical outcome. These findings support the conclusion that, at least in extensive-stage SCLC, established cell lines are not derived from patients whose tumors have an inherently different biology. Thus, the biologic fea-

tures of the cell lines may be representative of SCLC tumors as a whole. In addition, in vitro chemosensitivity testing of human lung cancer lines shows patterns of resistance reminiscent of what is observed in the clinic [192]. Cell lines from untreated SCLC patients were more sensitive to doxorubicin, carmustine, cisplatin, melphalan, vincristine, and etoposide than non-small cell lung carcinoma (NSCLC) cell lines. Cell lines from previously treated SCLC patients had drug sensitivity more similar to NSCLC lines. Consequently, it is reasonable to consider that these cell lines may be appropriate surrogates for primary patient material in the study of drug resistance mechanisms in human lung cancer. Several studies employing both primary tumor tissue and established cell lines have addressed the mechanisms of drug resistance in SCLC. As already mentioned, the phenotype of pleiotropic or multidrug resistance has been associated in some instances with the overexpression of a membrane glycoprotein which is able to actively transport multiple drugs outside the cell and is encoded by the A4DRl gene [186-1891. The role of this gene in lung cancer was explored by quantitating the level of expression of MDRl-specific message in mRNA extracted from a panel of lung cancer cell lines and tumors. The level of expression was found to be elevated in only 4/23 SCLC cell lines and 3/6 SCLC primary tumors studied. In no instances were elevations of MDRl expression to the very high levels sometimes noted in colon cancer found. Furthermore, no correlation was observed in SCLC material between MDRl expression and whether tumor material was derived from previously treated or untreated patients or from untreated patients who did or did not respond to initial chemotherapy. There was also no correlation between mRNA levels and in vitro chemosensitivity [193]. It would thus appear that enhanced drug efflux through ~170 glycoprotein is not a major factor in the clinical drug resistance of SCLC, and that other mechanisms must be operative. A clinical pilot study attempting to pharmacologically modify putative drug resistance resulting from MDRI overexpression in SCLC has already been performed [ 1941. The discovery that the high-level expression of the placental form of the drug metabolizing enzyme glutathione-4transferase correlated with a phenotype of pleiotropic drug resistance distinct from drug accumulation defects induced in the MCF-7 breast cancer cell line in vitro has several important implications [ 1901. Since this enzyme can not only detoxify a wide range of xenobiotics but also has its expression induced by exposure to these compounds, it was predicted that tumors arising in the context of long-term exposure to environmental carcinogens would be likely to have activated this detox-

123

ifying enzyme system. Since many drugs in common use are derivatives of natural products which can be detoxified by this mechanism, this provided a general framework within which many therapeutic failures could be explained. In fact, it has been shown that the level of glutathione-S-transferase enzymatic activity was increased in the lung tissue of smokers and that this was proportional to the cumulative tobacco exposure measured in pack-years [195]. There are several reports indicating the frequent high-level expression of this enzymatic system in NSCLC tumors and cell lines [196 2001. In contrast, SCLC tissues and cell lines consistently express lower levels of glutathione-S-transferase mRNA [ 1961, immunoreactivity [200], and enzymatic activity [197]. In a study of 15 SCLC cell lines, no correlation was found between glutathione-S-transferase enzymatic activity and sensitivity to doxorubicin or melphalan. Furthermore, there were no differences between cell lines derived from untreated patients when compared to those from previously treated patients [197]. In the aggregate, these observations support the view that the glutathione-S-transferase system does not explain the less common de novo or usual acquired resistance to chemotherapy observed in SCLC. Recent evidence points to alternative mechanisms of pleiotropic drug resistance which may be operative in SCLC. An SCLC cell line made resistant in vitro by graded exposure to doxorubicin was found to show collateral resistance to vincristine, vindesine, etoposide, cisplatin and X-irradiation, but not to melphalan, colchicine and actinomycin D. This drug resistance phenotype was explained on the basis of a combination of decreased intracellular levels of doxorubicin, increased DNA repair, and reduced DNA topoisomerase II activity [201, 2021. Since it appears that the repertoire of known drug resistance mechanisms used by SCLC is limited, further definition of these and other as yet unknown mechanisms is indicated in an effort to identify clinically relevant resistance pathways which might be modulated to improve the efficacy of existing chemotherapeutic agents. Examples of in vitro modulation of drug sensitivity in SCLC cell lines with cyclosporine and docosahexaneoic acid have been reported [203, 2041.

biogenic amine synthetic enzyme L-DOPA decarboxylase) [205]. The most striking manifestation of this phenotype is the elaboration of a large number of peptide hormones and growth factors, several of which were first identified as neuropeptides (see Table 1). This list continues to expand, the most recent additions being atria1 natriuretic factor [lo] and multiple opioid peptides [206]. The first peptide to be specifically documented to be produced by SCLC was gastrin-releasing peptide (GRP), the mammalian homologue of the amphibian tetradecapeptide bombesin [207, 2081. The details of this discovery are of historical interest. It had been recognized since the 1940s that the normal human bronchial

TABLE I Some hormones

produced

Product

Hormone

GRP [207,208,279]

+

IGF-1 [224]

+

Transferrin [225]

+

Calcitonin

+

[280]

AVP [28 I]

IV-B.I.

as a

Growth factors

The neuroendocrine nature of SCLC has been long recognized on morphological grounds (presence of neurosecretory granules), as well as biochemical grounds (expression of chromogranin A and the key

expressed

by human

small

produced

Receptor

expressed

+

Opioid peptides:

[206,

2821 /I-endorphin

+

enkephalin

+

dynorphin Neurotensin Glucagon

[280]

+

Substance

P [209]

+ + _

Somatostatin

+ + + +

+ +

[280,283]

[280,284]

CCK [226, 2841 TGFa: [280]

7 ? + +

VIP [28 I, 2841

+ _

ACTH

2811

+

?

GGAP

[285,286]

+

+

+ ?

1

?

+

ANF Galanin

[226]

Bradykinin

[226]

[ 1601

Interferon-y GM-CSF

[I601

+

+

?

+

?

+

Neurokinin

A [287]

?

Neurokinin

B [287]

?

+ +

?

+

?

+ ?

Acetylcholine,

musca-

rinic [288]

IV-B. Interruption of autocrine growth stimulation target for developmental therapeutics

and receptors

cell lung cancer

Acetylcholine,

nicotinic

VW Serotonin Adapted

[210]

+

from Ref. 289. Hormones

be involved in autocrine is assumed

growth

in boldface

regulatory

if shown by direct radioligand

cal effect of the cognate tion of a specific growth.

hormone

signal

have been shown

loops. Receptor

to

expression

binding

assay or if a biologi-

is demonstrated

by virtue of activa-

transduction

pathway

or stimulation

of

124

epithelium exhibited a cellular component whose morphological features were similar to that of a proposed diffuse neuroendocrine system [209]. Carcinoid tumors and SCLC shared many of these features, including the production of the biogenic amine serotonin [210]. However, the demonstration that these cells could also elaborate biologically active peptides remained elusive. The search for biologically active peptides throughout the animal world, particularly in the Bombina family of amphibians by Vittorio Erspamer, had generated a large family of candidate hormones. Using specific antibodies raised against the amphibian peptide bombesin, Wharton et al. demonstrated in 1978 the presence of immunoreactivity in the normal neuroendocrine cells of the human fetal lung [211]. The presence of bombesin-like immunoreactivity was subsequently reported in SCLC by several groups [207, 2081 and was later shown to be due to the expression of the mammalian homologue GRP [212]. GRP acts as a growth factor for normal human bronchial epithelial cells [213] and for SCLC both in vitro [214-2161 and in vivo [217]. The ability of the 2All monoclonal antibody raised against the carboxyl terminus of bombesin to block binding of GRP to its receptor led to the use of this reagent to study the impact of GRP on SCLC biology. It was found both in vitro and in vivo that exposing SCLC cells to this antibody deprived them of the trophic effect of the endogenously produced GRP and resulted in growth inhibition of the tumor cells [214]. These observations were the first demonstration that autocrine growth stimulatory loops might be an important factor in the genesis or maintenance of these malignant cells. Several synthetic peptides with antiGRP activity have been described (see Table 2), the most recent generation of which can block GRP binding at concentrations in the nanomolar range [218, 2191.

TABLE 2 Bombesin antagonists with in vitro activity against small cell lung cancer Structure (D-Argl, D-Pro*, D-TqP, Leu”) substance P (2201 (D-Argl, D-Phe5, D-Trp7.g, Leu”) substance P [220] (Leu’3-Y-CH2NH-Leu14) bombesin [221] n-Acetyl-GRP-20-26OCH2CH3 [218] (D-Phe6, Leu13-Y-CH2NHPhe14)bombesin (614) [219] Adapted from Ref. 289.

KS0 for growth

I& for binding

82 /LM 24 FM 1fiM

80.4 nM 4nM 3.1 nM

Several have been shown to inhibit SCLC growth in vitro [220,221]. This developmental work has generated novel compounds which hold promise as clinically useful anti-GRP drugs, and the original 2All antibody is currently undergoing human trial [222,223]. However, as is graphically documented by Table 1, GRP is only one of a multitude of peptides with potential growth-regulatory roles in SCLC. Both insulin-like growth factor I and transferrin-like growth factor participate in autocrine growth regulatory loops [224, 2251. In addition, individual SCLC ceil lines can be shown to be responsive to a number of distinct hormones. By monitoring the response of the intracellular calcium concentration, Woll and Rozengurt [226] showed that the SCLC cell line NCI-H345 could respond to eight different individual hormones (acetylcholine, ACTH, bradykinin, cholecystokinin, galanin, GRP, neurotensin, and vasopressin). If one also considers the factors known to be produced by SCLC which do not elicit a rise in intracellular calcium concentration, it becomes obvious that the stimulatory effects on the growth of SCLC cells by themselves (autocrine) or neighboring cells (paracrine) may be extremely varied and complex. In this setting, it would appear unlikely that a therapeutic strategy aimed at any one growth factor would have much clinical impact and that any serious attempt at interrupting these trophic effects must take into account this heterogeneity. Such strategies could be based on the use of combination of antibodies and peptide antagonists (each with their individual specificity) or on the use of peptide antagonists with broader specificities such as the substance P analogues reported to block the mitogenie effects of GRP, vasopressin, and bradykinin in Swiss 3T3 cells [227]. Alternatively, strategies based on interrupting intracellular signal-transducing events shared by these many hormones might be viable. For instance, enzyme effecters such as phospholipase C (PLC), protein kinase C (PKC), G-proteins coupling several receptors to their effecters, and membrane calcium channels are all actually or potentially amenable to pharmacologic manipulation. It is provocative that certain compounds already in common use in medical oncology have pleiotropic mechanisms of action which include modifications of signal transduction pathways. For example, the antiestrogen tamoxifen can block calcium channels [228], and the anti-tumor antibiotic doxorubicin is also an inhibitor of PKC 12291. In addition, the only known animal model for pulmonary neuroendocrine neoplasia appears to be sensitive to such manipulations. Schuller et al. have recently described a protocol wherein hamsters receive repeated injections of the carcinogen N-nitrosodiethylamine [230]. Under standard laboraty conditions, the animals develop peripheral

125

pulmonary adenocarcinomas within 20 weeks. However, when kept in a hyperoxic atmosphere of 60% oxygen, they develop pulmonary neuroendocrine tumors within 8 weeks. Remarkably, the administration of a calcium/ calmodulin inhibitor prevents the appearance of the neuroendocrine tumors but not the adenocarcinomas [23 11. IV-B.2. Gastrin-releasing peptide stimulated signal transduction pathways

The above observations suggest that the manipulation of signal transduction pathways may prove to be a fertile ground for the developmental therapeutics of lung cancer. To better define potential targets, efforts to map the intracellular events following exposure of SCLC to the mitogen GRP are underway. GRP stimulation of SCLC is associated with the activation of a major signal transduction system centered on the enzyme PLC. In recent years, receptors activating PLC have been found to be coupled to it through the action of other membrane-associated proteins characterized by the ability to bind guanine nucleotides in a reversible manner [232] (see Fig. 3). These guanine-nucleotide binding proteins or G-proteins are molecular switches which transmit and amplify the signal initiated by the ligand-bound receptor to an effector, in this case PLC. The ligand-bound receptor adopts a conformational state which causes the binding of GTP to the G-protein. The GTP-bound G-protein is then able to stimulate the effector. The system can revert to baseline because of the intrinsic ability of the G-protein to hydrolyze the bound GTP and thus return to an inactive state. The function

of some G-proteins can be modulated through phosphorylation by the enzyme PKC [233] or through a posttranslational modification (ADP-ribosylation) catalyzed by certain bacterial toxins (cholera and pertussis toxins) which have been used as probes for the detection of Gprotein involvement in biological responses. A hallmark of receptor-effector coupling by a G-protein is the ability of non-hydrolyzable GTP analogs, which cause an irreversible activation of the G-protein, to modulate the affinity of receptor binding and the intensity of the transduced signal such as PLC activity. Bacterial toxins induce the same modifications when the involved G-protein is a substrate for their action (reviewed by Spiegel, Ref. 234). PLC activation results in the hydrolysis of an integral membrane phospholipid, phosphatidylinositol bisphosphate (PIP$ to form two products, inositol trisphosphate (IPs) and diacylglycerol (DAG). IPs in turn binds to a receptor on the endoplasmic reticulum, stimulating the release of endogenous calcium stores and transiently increasing the intracellular concentration of free calcium ([Ca*+]ir,) [235]. DAG activates the calcium and phospholipid-dependent protein kinase PKC. PKC activation can be followed by the activation of other pathways, such as the membrane Na+/H+ ‘antiporter’, causing an elevation of intracellular pH, or the transcriptional activation of certain genes such as the protooncogenes c-fos, c-&n and c-myc [236, 2371. After stimulation by a PLC-activating ligand, cells are often transiently refractory to a repeat stimulation by the same (homologous desensitization) or other (heterologous desensitization) ligands. PKC has been implicated in some of these regulatory events. Fig. 4 summarizes work which has shown that GRP

PMA

Q+y+y c-fos

c-myc

Fig. 3. Intracellular of phospholipase nucleotide ylinositol

binding

pathways protein;

bisphosphate;

lular concentration

involved in receptor

C. Abbreviations

PLC, phospholipase

IP3, inositol

of free calcium;

mediated

used are: R, receptor;

C; PIPZ, phosphatid-

trisphosphate; DAG,

tein kinase C.

activation G, guanine-

[Ca2+]i, intracel-

diacylglycerol;

PKC,

pro-

Fig. 4. Intracellular

pathways

small cell lung cancer with the addition

activated

by gas&in-releasing

cell lines. Abbreviations

of CT, cholera

toxin; and PMA.

istate 13-acetate.

peptide in

used are as in Fig. 3. phorbol

12-myr-

126

stimulation activates PLC in SCLC, both in metabolically labeled whole cell and membrane preparations [238, 2391. This activation is accompanied by an elevation of [Ca2+]irree [240], but PKC response has not yet been characterized. However, GRP stimulation of IP3 formation and increase in [Ca2+]irree were inhibited by prior treatment with the PKC activator phorbol 12myristate 13-acetate (PMA), suggesting that PKC might exert negative feedback regulation on this response.

a complete inhibition of the growth of three SCLC cell lines bearing the GM 1 ganglioside receptor for CT. The effects on signal transduction preceded the effects on cellular metabolic activity, suggesting that CT may be acting principally by depriving the cell of the trophic effects of multiple growth factors [241] and providing a model as well as a potential target for the therapeutic manipulation of growth-associated signal transduction pathways in SCLC.

IV-B.3. Efects of cholera toxin on signal transduction pathways andgrowth Because non-hydrolyzable GTP analogues could modulate PLC activation in response to GRP, it was concluded that the GRP receptor in SCLC was coupled to PLC by a G-protein [239]. The nature of this putative G-protein was investigated further by studying the effects of G-protein modifying bacterial toxins on GRPmediated signals in SCLC. Cholera toxin (CT) is a multimeric bacterial protein composed of five jI subunits, which bind to a specific receptor, the cell surface glycolipid GM1 ganglioside, and one CIsubunit which enters the cell. The a subunit is an enzyme which catalyzes the transfer of ADP-ribose from NADP to certain G-proteins, thus modifying their function by abolishing their intrinsic GTPase activity. CT was found to inhibit GRP-stimulated generation of IP3 and increase of [Ca2+]ir, in the SCLC cell line NCI-H345 [238]. The best characterized action of CT is to modify the G-protein coupled to adenylate cyclase and stimulate the formation of intracellular cyclic AMP (CAMP). CAMP is itself an important second messenger able to activate a family of CAMP-dependent protein kinases mediating pleiotropic cellular effects. Because of this, the possibility that the effects of CT on GRP responses might be indirectly caused by an elevation of CAMP was considered. However, preincubation of SCLC with cyclic AMP analogues had no effect on the generation of GRP-stimulated signals [238]. These observations initially suggested that the GRP receptor of SCLC might be coupled to PLC by a CT-sensitive G-protein. The effects of CT on the signal transduction pathways and biology of SCLC have recently been studied in more detail [241]. CT pretreatment led to an inhibition of signal transduction generated not only by GRP but also by vasopressin and fetal calf serum. This was accompanied by a loss of cell surface membrane ruffling and a decrease in GRP-stimulated PLC activity in membranes prelabeled with [3H]inositol, but with no change in GRP receptor number or affinity. This indicated that CT could perturb membrane function related to PLC activity but not through an effect on the G-protein coupling the GRP receptor to PLC. More importantly, CT led to

IV-C. Genetic lesions in SCLC We now appreciate that established SCLC harbors a wide range of somatic genetic abnormalities, including activation of dominant oncogenes as well as inactivation of several newly recognized tumor suppressor genes. These data have been extensively reviewed recently [242] and will be only summarized here. The appreciation of the range of genetic lesions present in human SCLC followed from a number of serendipitous observations, as well as from specific studies of a panel of SCLC cell lines in the context of the historical development of the field of oncogene research. It was clear early on that SCLC tumors had suffered a number of genetic events. Early cytogenetic studies revealed abnormalities of virtually every chromosome, with the most consistent change being that of a deletion of the

TABLE Major

3 dominant

oncogenes

and tumor

suppressor

genes altered

in

small cell lung cancer Gene

Frequency*

Function

Dominant ras family

O/42 cell lines [272]

~21 membrane-associated protein.

Signal transduc-

tion? Activated

by point

mutations.

myc family

Nuclear

Amplified

phosphoproteins.

Function expression

unknown.

c-myc

coupled

to cell

11-24s

increased Tumor

of and

[262,

Overex-

pressed in 89% of cell

with IYJSin transforming cells. Activated

of tumors

264,267,268].

cycle. All can cooperate primary

in 30-50s

cell lines [264,268]

by

lines and 83% of tumors [268].

expression.

sup-

pressor 3P

Putative

rb

genes not yet identified. Nuclear phosphoprotein.

tumor

P53

cell cycle regulation. Nuclear protein. ? function.

92-100% [250]

suppressor ?

Inactivated

[253]. a Reference

numbers

are in parentheses.

in > 90%

[252, 254, 2901. Inactivated in > 50%

127

short arm of chromosome 3 [243,244]. Further cytogenetic studies as well as restriction fragment length polymorphism analysis confinned the presence of multiple areas of loss of genetic information, particularly at 3p (in 92-100s of SCLC) [245-2501, 1lp (the locus of the Wilm’s tumor gene) [251], 13q (the locus of the retinoblastoma gene), and 17p (the locus of the ~53 gene) [249]. The retinoblastoma gene and ~53 have functional characteristics of tumor suppressor genes, and both have been shown to be inactivated in a high proportion of SCLC by diverse mechanisms including deletions, lack of expression and point mutations [252-2561. It is in this context that it is anticipated that at least one more tumor suppressor gene is localized on 3p; its identification and molecular cloning is the focus of intense efforts in several laboratories. In addition, the recent cloning of a candidate for the Wilm’s tumor gene [257] will allow the study of its expression in SCLC and other tumors which show losses at 1lp. The recent observation that mutant forms of the ~53 protein are more stable and can be detected by standard immunohistochemical techniques [258J is bound to facilitate the analysis of their clinical and biological significance. In contrast to the above genes whose involvement in carcinogenesis is through loss of their normal function in the restrictive control of cellular growth, several other genes manifest themselves by an inappropriate and deregulated functional activation. A paradigm for these dominant oncogenes in SCLC has been the myc family of genes. The genes c-myc, N-myc and L-myc all encode nuclear phosphoproteins whose precise functions remain to be defined. Because they can all cooperate to varying degrees in transforming primary rodent cells, they are thought to stimulate or enhance cellular growth [2592611. All three genes have been shown to be overexpressed in some SCLC tumors and cell lines by virtue of genomic amplification (in 10-20s of primary tumors) or deregulated transcription [262-2691. Overexpression of a myc family gene appears to confer more aggressive biologic properties to SCLC, as tumors with this property are more tumorigenic in nude mice [266] and the patients from whom they are derived have a worse prognosis [263, 2641. There is a relative paucity of information on the expression and function of the scores of other dominant oncogenes that have been described. In particular, the ras family of oncogenes, which can be activated by well characterized point mutations, has been shown to be activated in about 20% of NSCLC, seemingly uniquely at codon 12 of c-ki-rus [270,271]. We have recently studied rus mutations in a panel of SCLC and NSCLC cell lines and have found that 23/63 NSCLC cell lines had rus

gene mutations, most frequently involving codon 12 of c-k&as but also other codons of two other ras family genes. In contrast, none of 42 SCLC cell lines had mutations of any ras gene at any of the three codons studied [272]. These observations suggest that rus mutations do not play an active role in the pathogenesis of SCLC. However, the introduction of an activated rus gene into SCLC cell lines has been associated with the loss of neuroendocrine features, morphological transformation towards an NSCLC histology, and the appearance of growth factors and growth-factor receptors more commonly associated with NSCLC [273,274]. It is not clear whether these provocative observations are relevant to the natural history of lung cancer. As knowledge of the genetic lesions present in SCLC expands, a number of themes are emerging. Any given tumor usually exhibits multiple abnormalities in both the tumor suppressor genes and the dominant-acting oficogenes. In fact, several cell lines harbor abnormalities of all three defined or putative tumor suppressor genes recognized to be involved in SCLC (3p, rb, and p53), as well as lesions at other chromosomal loci combined with a deregulated myc gene [253]. This knowledge may eventually be exploitable in the clinic. Analysis of the cumulative effect of similar genetic abnormalities (termed an allelotype by Vogelstein et al., Ref. 275) is of prognostic value in colon cancer and may be useful in SCLC in terms of targeting therapy to biologically defined subsets of patients. In addition, one can envision strategies designed to deprive tumor cells of the function of a constitutively activated oncogene [276], to replace a critically missing function through gene therapy, or to target novel treatments at mutated genes or their products [277,278].

V. Conelusions

The development of more effective therapeutic approaches to SCLC is at a crossroads. In no other area of oncology is the dialogue between clinicians and molecular and cellular biologists more important and likely to bear fruit. The plateau in our recent therapeutic achievements must not discourage continuing to address the incrementally progressive but important issues of determining the optimal dose and schedule of the agents, including radiation therapy, currently available. It is hoped that the drug screening program utilizing panels of human lung cancer cell lines will identify useful new drugs. However, we must not be timid in exploring the new possibilities identified by our rapidly expanding knowledge of the basic biology of SCLC.

128

References

DH. Combined Oncol7:

1 Matthews

MJ, Kanhouwa

of residual surgical

S, Pickren

and metastatic

resection

tumor

for lung cancer.

Cancer

20 American

D. Frequency

undergoing

Chemother

Rep (Part 3)

AS, et al. Small cell lung cancer

and recent obstacles.

Int J Radiat

Oncol

JD, Pass H, Glatstein

De Vita VT, Hellman and Practice 4 Silverberg

E, Boring

of surgery

CC, Squires

JG. Medical

c,*rcinoma

RA, Hun

7 Lanzotti

Research

Council

comparative

treatment Lancet

ii: 63, 1973.

ME. Alkylating

agents

Boyle

LE,

Smith

Gehan

Samuels ML. Survival

with inoperable

based on simple clinical criteria.

EA,

MH,

Bemath

JD. Serial fiberoptic

for small cell carcinoma

AM,

Matthews

bronchoscopy

MJ, Bunn

during

39: PA,

of patients

at

high risk of relapse. Chest 74: 53 1, 1978. 9 Bunn PA, Ridgway VT, Hellman

EC. Paraneoplastic

S, Rosenberg

tice of Oncology.

SA, eds. Cancer:

Philadelphia:

lung cancer

HJ, Lambert

V. Antagonism

gene in small cell Inst 82:

EH, Griesmann calcium

with and without

by autoantibodies,

GE, Olivera channels

B, Lennon

in small cell car-

Lambert-Eaton

o-conotoxin

myasthenic

and adenosine.

Cancer

Health

histological

Organization.

typing

The

World

of lung tumours:

Health

Second

Organization

edition.

Am J Clin

B, Wright

SE, Moatamed

cell lung carcinoma: 14 Burdon

JGW,

F. Electron

Clinical correlation.

Sinclair

of the lung: Prognosis

RA, Henderson in relation

microscopy

J Clin Onco12:

in small 910, 1984.

MM. Small cell carcinoma

to histologic

subtype.

Chest 76:

DN, Matthews

subtype

to therapy,

according

of histologic

of the lung on clinical

and survival.

presenta-

J Nat1 Cancer

Inst 65:

K, Hansen

subtyping

to the classification

HH. The prognostic

of small cell carcinoma of the World

Health

significance of the lung Organization.

52: 2144, 1983.

17 Radice PA, Matthews MJ, Ihde, DC, et al. The clinical behavior of ‘mixed’ small cell/large cell bronchogenic carcinoma compared to ‘pure’ small cell subtypes. 18 Aisner

SC, Finkelstein

deschel JC, Eggleston phology 1990. 19 Mangum

procedures

D, Maurer

of the

of patients

in staging

factors

proce-

over 14 years.

EP. The relative value

for developing

lung cancer.

prognostic

mo-

J Clin Oncol8:

Clin Oncol7:

LH, Ware JH, et al. Prognostic of the lung: An analysis

2047.

GA, Matthews

section

in the management

Thorac

Cardiovasc

27 Johnson

factors

in

of 1,521 patients.

J

344, 1989.

DC. Women

MJ, Keehn

RJ. Surgical

of small cell carcinoma

Surg 84: 48

BE, Steinberg

re-

of the lung. J

1, 1982.

SM, Phelps R, Edison

small cell lung cancer

28 Plsterlind cancer:

K, Andersen

patients

PK. Prognostic

Multivariate

chemotherapy

model

based

with or without

29 Souhami

RL, Bradbury

bias JS. Prognostic

M, Veach SR, Ihde live longer than men.

factors

in small cell lung

on 778 patients

irradiation.

Cancer

treated

with

Res 46: 4189,

ured at diagnosis

small-cell

I, Geddes DM, Spiro SG, Harper

significance

DM,

DS, Abeloff

of the lung.

FA, Hainsworth

MD,

Ruck-

of variant-mor-

J Clin Oncol

JD, Hande

PG. To-

parameters

meas-

of the lung. Cancer

Res

45: 2878, 1985. E, Finkelstein

small-cell

lung cancer

Oncol4:

H, Wolkove

N, Kreisman

on protocol:

Potential

H. Treatment

of

bias of results. J Chn

1314, 1986. of small cell lung cancer:

tive on two decades of clinical research. 32 Johnson

DH, Greco

FA. Small cell carcinoma

Crit Rev Oncol/Hematol4: 33 Edmonson

A perspec-

Semin Oncol 15: 278, 1988. of the lung. CRC

303, 1986.

JH, Lagakos

SW, Selawry

in the treatment

ma and adenocarcinoma

OS, et al. Cyclophospha-

of inoperable

of the lung. Cancer

small cell carcinoTreat

Rep 60: 925,

1976. 34 Lowenbraun

S, Bartolucci

agent chemotherapy 1979.

A, Smalley RV, Lynn M, Krauss of combination

in small cell lung carcinoma.

35 Albert0 P, Brunner KW, Treatment of bronchogenic quential

combination

clophosphamide,

chemotherapy

S, Du-

over single

Cancer

44: 406,

Martz G, Obrecht JP, Sonntag RW. carcinoma with simultaneous or se-

chemotherapy,

procarbazine

including

and vincristine.

methotrexate, Cancer

cy-

38: 2208,

1976.

50: 2894, 1982.

Ettinger

JC. The clinical significance

carcinoma

MD, Greco

Cancer

of laboratory

in small cell carcinoma

rant JR. The superiority

FR, (dsterlind

of histopathologic Cancer

MJ, Ihde DC, et al. Influence

of small cell carcinoma

tion, response 1225, 1980. 16 Hirsch

on prognostic

small-cell

carcinoma

mide and CCNU

302, 1979. 15 Camey

An analysis

data base. J Clin Oncol

Effects of changes

31 Seifter EJ, Ihde DC. Therapy

Path01 77: 123, 1982. 13 Copple

25 Spiegelman

30 Quoix

Res 48: 4719, 1988. 12 World

RB. Determinants

Am J Med 85: 194, 1988.

AF, Johnson

cell lines. J Nat1 Cancer

of voltage-gated

of patients

syndrome

factor

re-

patients.

1986.

305, 1990. 11 DeAizpurua cinomas

and tumor

and Prac-

1989; 1896.

RI, Birrer MJ, Gazdar

of the atria1 natriuretic

tumors

In: De Vita

Principles

J.B. Lippincott,

10 Bliss DP, Battey JF, Linnoila BE. Expression

syndromes.

Group

J Clin Oncol8: 1042, 1990. 24 Sheehan RG, Balaban EP, Cox JV, Frenkel

26 Shields TW, Higgins

chemotherapy

of the lung: Early detection

M, Livingston

SM, Phelps R, et al. Outcome

technology

dels in extensive-stage

P. Influence

on the long-term

589, 1986.

with small cell lung cancer:

staging

of Can-

A study of 150 operable

Oncology

8: 1563, 1990. 23 Dearing MP, Steinberg

small-cell

303,1977. 8 Ihde DC, Cohen

HH, Dombernowsky

in small cell lung cancer:

Southwest

In: Beahrs 0,

for Staging

1990.

lung cancer: An integration Cancer

Lung cancer.

to chemotherapy

JJ, LeBlanc

outcome

of conventional

TL,

prior

Clin Onco122:

dures and imaging

of small-celled

of prognostic

Minna

trial

Am J Med 46: 516, 1969.

DR,

variables

1990. CA

M, Hansen

KS, Crowley

2,580-patient

of the bronchus.

VJ, Thomas

22 Albain

1989; 591.

statistics

J Clin

1988; 115.

sults in small cell lung cancer:

J.B. Lippincott,

TS. Cancer

on Cancer.

J.B. Lippincott,

resection

of improved

for primary

carcinoma.

of the lung, In:

cell lung cancer.

R, Myers M, eds. Manual

K, Hansen

of surgical

Principles

>hrey E, Close H, Patno

in bronchogenic

cer. Philadelphia:

SA, eds. Cancer:

Philadelphia:

and radiotherapy

or oat-celled 6 Green

E, Ihde DC. Cancer

S, Rosenberg

of Oncology.

40: 9, 1990. 5 Fox W, Scadding

D, Hutter

Eur J Cancer

Biol Phys 10: 515, 1984. 3 Minna

Joint Committee

Henson

curative

21 Bsterlind

4: 63, 1973. 2 Morstyn G, Ihde DC, Lichter 1973-1983: Early progress

J, Robinette

in patients

small cell and non-small

607, 1989.

8: 402,

KR, Johnson

36 Bleehan NM, Fayers PM, Girling DJ, Stephens RJ. Survival, adverse reactions and quality of life during combination chemotherapy compared with selective palliative cancer. Respir Med 83: 5 I, 1989. 37 Jackson

treatment

for small-cell lung

DV, Case LD, Zekan PJ, et al. Improvement

of long-term

129 survival in extensive small-cell lung cancer. J Clin Oncol 6: 1161, 1988. 38 Jett JR, Everson L, Themeau TM, et al. Treatment of limitedstage small-cell lung cancer with cyclophosphamide, doxorubicin, and vincristine with or without etoposide: A randomized study of the North Central Cancer Treatment Group. J Clin Oncol 8: 33, 1990. 39 Lowenbraun S, Birch R, Buchanan R, et al. Combination chemotherapy in small cell lung carcinoma: A randomized study of two intensive regimens. Cancer 54: 2344, 1984. 40 Hong WK, Nicaise C, Lawson R, et al. Etoposide combined with cyclophosphamide plus vincristine compared with doxorubicin plus cyclophosphamide plus vincristine and with high-dose cyclophosphamide plus vincristine in the treatment of small-cell carcinoma of the lung: A randomized trial of the Bristol Lung Cancer Study Group. J Clin Oncol7: 450, 1989. 41 Slevin ML, Clark PI, Joel SP, et al. A randomized trial to evaluate the effect of schedule on the activity of etoposide in small-cell lung cancer. J Clin Oncol7: 1333, 1989. 42 Mead GM, Thompson J, Sweetenham JW, Buchanan RB, Whitehouse JMA, Williams CJ. Extensive stage small cell carcinoma of the bronchus: A randomized study of etoposide given orally by one-day or five-day schedule together with intravenous adriamycin and cyclophosphamide. Cancer Chemother Pharmacol 19: 172, 1987. 43 Evans WK. Osoba D, Feld R, Shepherd FA, Bazos MJ, DeBoer G. Etoposide (VP-16) and cisplatin: An effective treatment for relapse of small-cell lung cancer. J Clin Oncol 3: 65, 1985. 44 Evans WK, Shepherd FA, Feld R, Osoba D, Dang P, DeBoer G. VP-16 and cisplatin as first-line therapy for small-cell lung cancer. J Clin Oncol 3: 1471, 1985. 45 Wolf M, Havemann K, Holle R, et al. Cisplatin/etoposide versus ifosfamide/etoposide combination chemotherapy in small-cell lung cancer: A multicenter German randomized trial. J Clin Oncol 5: 1880, 1987. 46 Ihde DC, Mulshine JL, Kramer BS, et al. Randomized trial of high vs. standard dose etoposide and cisplatin in extensive stage small cell lung cancer (Abstr.). Proc Am Sot Clin Oncol 10: 240, 1991. 47 Carney DN, Grogan L, Smit EF, Harford P, Berendsen HH, Postmus PE. Single-agent oral etoposide for elderly small cell lung cancer patients. Semin Oncol 17 (Suppl. 2): 49, 1990. 48 Bork E, Hansen M, Dombernowsky P. Hansen SW, Pedersen AG, Hansen HH. Teniposide (VM-26), an overlooked highly active agent in small-cell lung cancer: Results of a Phase II trial in untreated patients. J Clin Oncol4: 524, 1986. 49 Clark PI, Cottier B, Joel SP, Thompson PI, Slevin ML. Prolonged administration of single-agent oral etoposide in patients with untreated small cell lung cancer (Abstr.). Proc Am Sot Clin Oncol 9: 226, 1990. 50 Cohen MH, Creaven PJ, Foss&k BE, et al. Intensive chemotherapy of small cell bronchogenic carcinoma. Cancer Treat Rep 61: 349, 1977. 5 1 Mehta C, Vogl SE. High-dose cyclophosphamide in the induction therapy of small cell lung cancer: Minor improvements in rate of remission and survival (Abstr.). Proc Am Assoc Cancer Res 23: 155, 1982. 52 Souhami RL, Finn G, Gregory WM. et al. High-dose cyclophosphamide in small cell carcinoma of the lung. J Clin Oncol 3: 958, 1985. 53 Figueredo AT, Hryniuk WM, Strautmanis I, Frank G, Rendell S. Co-trimoxazole prophylaxis during high-dose chemotherapy of small-cell lung cancer. J Clin Oncol 3: 54, 1985.

54 Johnson DH, Einhorn LH, Birch R, et al. A randomized comparison of high-dose versus conventional-dose cyclophosphamide, doxorubicin, and vincristine for extensive-stage small-cell lung cancer: A Phase III trial of the Southeastern Cancer Study Group. J Clin Oncol5: 1731, 1987. 5.5 Brower M, Ihde DC, Johnston-Early A, et al. Treatment of extensive stage small cell bronchogenic carcinoma: Effects of variation in intensity of induction chemotherapy. Am J Med 75: 993, 1983. 56 Abeloff MD, Ettinger DS, Order SE, et al. Intensive induction chemotherapy in 54 patients with small cell carcinoma of the lung. Cancer Treat Rep 65: 639,198l. 57 Valdivieso M, Cabanillas F, Keating M, et al. Effects of intensive induction chemotherapy for extensive-disease small cell bronchogenie carcinoma in protected environment-prophylactic antibiotic units. Am J Med 76: 405, 1984. 58 Johnson DH, DeLeo MJ, Hande KR, Wolff SN, Hainsworth JD, Greco FA. High-dose induction chemotherapy with cyclophosphamide, etopodise, and cisplatin for extensive-stage small-cell lung cancer. J Clin Oncol5: 703, 1987. 59 Wolff SN, Birch R, Sarma P, Greco FA. Randomized dose-response evaluation of etoposide in small cell carcinoma of the lung: A Southeastern Cancer Study Group trial. Cancer Treat Rep 70: 583, 1986. 60 Goldie JH, Coldman AJ, Gudauskas GA. Rationale for the use of alternating non-cross resistant chemotherapy. Cancer Treat Rep 66: 439, 1982. 61 Bsterlind K, Sorenson S, Hansen HH, Dombernowsky P, Hirsch FR, Hansen M, Rerth M. Continuous versus alternating combination chemotherapy for advanced small cell carcinoma of the lung. Cancer Res 43: 6085, 1983. 62 Evans WK, Feld R, Murray N, Willan A, Coy P, Osoba D, Shepherd FA, Clark D, Levitt M, MacDonald A, Wilson K, Shelley W, Pater J. Superiority of alternating non-cross-resistant chemotherapy in extensive small cell lung cancer. Ann Intern Med 107: 451. 1987. 63 Einhorn LH, Crawford J, Birch R, Omura G, Johnson DH, Greco FA. Cisplatin plus etoposide consolidation following cyclophosphamide, doxorubicin, and vincristine in limited small-cell lung cancer. J Clin Oncol6: 451, 1988. 64 Roth BJ, Johnson DH, Greco FA, Einhorn LH, Goodlow JL, Cherng N, Randolph JA, Schacter LP. A Phase III trial of etoposide and cisplatin versus cyclophosphamide, doxorubicin, and vincristine versus alternation of the two therapies for patients with extensive small cell lung cancer: Preliminary results (Abstr.). Proc Am Sot Clin Oncol8: 225, 1989. 65 Tamura T, Fukuoka M, Furuse K, Nishiwaki Y, Ikegami H, Saijo N, Suemasu K. Japanese multicenter randomized trial: Cyclophosphamide/adriamycin/vincristine (CAV) versus cisplatin/etoposide (PVP) versus CAV alternating with PVP in patients with small cell lung cancer (Abstr.). Proc Am Sot Clin Oncol8: 220, 1989. 66 Feld R, Evans WK, DeBoer G, Quirt IC, Shepherd FA, Yeoh J, Pringle J, Payne D, Herman J, Chamberlain D, Brown T, Baker M, Myers R, Blackstein M, Pritchard K. Combined modality induction therapy without maintenance chemotherapy for small cell carcinoma of the lung. J Clin Oncol2: 294, 1984. 67 Spiro SG, Souhami RL, Geddes DM. Ash CM, Quinn H, Harper PG. Tobias JS, Partridge M, Eraut D. Duration of chemotherapy in small cell lung cancer: A Cancer Research Campaign trial. Br J Cancer 59: 578, 1989. 68 Ettinger DS, Finkelstein DM, Abeloff MD, Ruckdeschel JC, Aisner SC, Eggleston JC. A randomized comparison of standard chemotherapy versus alternating chemotherapy and maintenance

69

70

71

72

73

74

75

76

77

78 79

80

81 82 83 84

85

86

versus no maintenance therapy for extensive stage small-ceB lung cancer: A Phase III study of the Eastern cooperative Oncology Group. J Clin Oncol8: 230,199O. Bleehan NM, Fayers PM, Girling DJ. Stephens RJ. Controlled trial of twelve versus six courses of chemotherapy in the treatment of small cell lung cancer. Br J Cancer 59: 584,1989. Splinter TAW, EORTC Lung Cancer Cooperative Group. Induction vs. induction plus maintenance chemotherapy in small cell lung cancer: Definitive evaluation (Abstr.). Proc Am Sot Clin Oncol7: 202, 1988. Taylor CW, Crowley J, Williamson SK, Miller TP, Taylor SA, Shanker Giri TG, Stephens RL, Livingston RB. Treatment of small-cell lung cancer with an alternating chemotherapy regimen given at weekly intervals: A Southwest Oncology Group pilot study. J Clin Oncol8: 1811, 1990. Batist G, Camey DN, Cowan KH, Veach SR, Gilliom M, Bunn PA, Ihde DC. Etoposide (VP-16) and cisplatin in previously treated smaBce11 lung cancer: Clinical trial and in vitro correlates, J Clin Oncol4: 982, 1986. Giaccone G, Donadio M, Bonardi G, Testore F, Calciati A. Teniposide in the treatment of small-cell lung cancer: The influence of prior chemotherapy. J Clin Oncol6: 1264,1988. Johnson DH, Greco FA, Strupp J, Hande KR, Hainsworth JD. Prolonged administration of oral etoposide in patients with relapsed or refractory small-cell lung cancer: A Phase II trial. J Clin Oncol8: 1613,199O. Ihde DC, Deisseroth AB, Lichter AS, et al. Late intensive combined modality therapy followed by autologous bone marrow infusion in extensive stage small cell lung cancer. J Clin Oncol4: 1443, 1986. Humblet Y, Symann M, Bosly A, et al. Late intensification chemotherapy with autologous bone marrow transplantation in selected small-cell carcinoma of the lung: A randomized study. J Clin Oncol5: 1864,1987. Spitxer G, Farha P, Valdivieso M, et al. High-dose intensification therapy with autologous bone marrow support for limited smallcell bronchogenic carcinoma. J Clin Oncol4: 4,1986. Livingston RB. Small-cell lung cancer: Whither late intensification? J Clin Oncol4: 1437, 1986. Issell BF, Einhom LH, Comis RL, et al. Multicenter Phase II trial of etoposide in previously treated small cell carcinoma of the lung. Cancer Treat Rep 69: 127,1985. Levenson RM, Ihde DC, Huberman MS, Cohen MH, Bunn PA, Minna JD. Phase II trial of cisplatin in small cell carcinoma of the lung. Cancer Treat Rep 65: 905, 1981. Smith IE. New treatments for small cell lung cancer. Chest 96 (Suppl.): 59, 1989. Aisner J. Identification of new drugs in small cell lung cancer: Phase II agents lirst? Cancer Treat Rep 71: 1131, 1987. Giaccone G. Identification of new drugs in pretreated patients with small cell lung cancer. Eur J Cancer Clin Oncol25: 411,1989. Cullen MH, Hilton C, Stuart NSA. Evaluating new drugs as first treatment in patients with small-cell carcinoma: Guidelines for an ethical approach with implications for other chemotherapy-sensitive tumors (letter). J Clin Oncol6: 1356, 1988. Cullen MH, Smith SR, Benfield GFA, Woodroffe CM. Testing new drugs in untreated small cell lung cancer may prejudice the results of standard treatment: A phase II study of oral idarubicin in extensive disease. Cancer Treat Rep 71: 1227, 1987. Blackstein M, Eisenhauer EA, Wierzbicki R, Yoshida S. Epirubitin in extensive small-cell lung cancer: A phase II study in previously untreated patients: A National Cancer Institute of Canada Clinical Trials Group study. J Clin Oncol8: 385, 1990.

87 Evans WK, Eisenhauer EA, Cormier, Y, et al. Phase II study of amonafide: Results of treatment and lessons learned from the study of an investigational agent in previously untreated patients with extensive small-cell lung cancer. J Clin Oncol8: 390, 1990. 88 Ettinger DS, Finkelstein DM, Abeloff MD, Bonomi PD. Justification for evaluating new anticancer drugs in selected untreated patients with a chemotherapy-sensitive advanced cancer: An ECOG randomized study (Abstr.). Proc Am Sot Clin Oncol9: 224, 1990. 89 Ettinger DS. Evaluation of new drugs in untreated patients with small-cell lung cancer: Its time has come. J Clin Oncol8: 374,199O. 90 Gaxdar AF, Steinberg SM, Russell EK, et al. Correlation of in vitro drug sensitivity testing results with response to chemotherapy and survival in extensive stage small cell lung cancer: A prospective clinical trial. J Nat1 Cancer Inst 82: 117,199O. 91 Tsai CM, Ihde DC, Kadoyama C, Venzon D, Gazdar AF. Correlation of in vitro drug sensitivity testing of long-term small cell lung cancer cell lines with patient response and survival. Eur J Cancer 26: 1148, 1991: _ 92 Boyd MR, Shoemaker RH, McLemore TL, et al. New drug development. In: Roth J, Ruckdeschel J, Weisenburger T, eds. Thoracic Oncology. Philadelphia: W.B. Saunders, 1989; 711. 93 Bunn PA, Ihde DC. Small cell bronchogenic carcinoma: A review of therapeutic results. In: Livingston RB, ed. Lung Cancer 1. The Hague: Martinus Nijhoff, 1981; 169. 94 Bunn PA, Lichter AS, Makuch RW, et al. Chemotherapy alone or chemotherapy with chest radiation therapy in limited stage small cell lung cancer: A prospective randomized trial. Ann Intern Med 106: 655,1987. 95 Perry MC, Eaton WL, Propert KJ, et al. Chemotherapy with or without radiation therapy in limited small-ceil carcinoma of the lung. N Engl J Med 316: 912,1987. 96 Perez CA, Einhom L, Oldham RK, et al. Random&d trial of radiotherapy to the thorax in limited small-cell carcinoma of the lung treated with multiagent chemotherapy and elective brain irradiation: A preliminary report. J Clin Oncol2: 1200, 1984. 97 Fox RM, Woods RL, Brodie GN, Tattersall MHN. A randomized study: Small cell anaplastic lung cancer treated by combination chemotherapy and adjuvant radiotherapy. Int J Radiat Oncol Biol Phys 6: 1083,198O. 98 Gsterlind K, Hansen HH, Hansen HS, Dombemowsky P, Hansen M, R&h M. Chemotherapy versus chemotherapy plus irradiation in limited small cell lung cancer: Results of a controlled trial with 5 years follow-up. Br J Cancer 54: 7,1986. 99 Kies MS, Mira JG, Crowley JJ, et al. Multimodal therapy for limited small-cell lung cancer: A randomized study of induction combination chemotherapy with or without thoracic radiation in complete responders; and with wide-field versus reduced-field radiation in partial responders: A Southwest Oncology Group study. J Clin Oncol5: 592, 1987. 100 Souhami RL, Geddes DM, Spiro SG, et al. Radiotherapy in small cell cancer of the lung treated with combination chemotherapy: A controlled trial. Br Med J 288: 1643, 1984. 101 Brooks BJ, Seifter EJ, Walsh TE, et al. Pulmonary toxicity with combined modality therapy for limited stage small cell lung cancer. J Clin Oncol4: 200,1986. 102 Arriagada R, Le Chevalier T, Baldeyrou R, et al. Alternating radiotherapy and chemotherapy schedules in small cell lung cancer, limited disease. Int J Radiat Oncol Biol Phys 11: 1461, 1985. 103 Camey DN, Mitchell JB, Kinsella TJ. In vitro radiation and chemotherapy sensitivity of established cell lines of human small cell lung cancer and its large cell morphological variant. Cancer Res 43: 2806, 1983. 104 Turrisi AT, Glover DJ, Mason BA. A preliminary report: Con-

131

current

twice-daily

therapy

for limited small cell lung cancer.

radiotherapy

plus platinum-etoposide

chemo-

Int J Radiat

Oncol Biol

Phys 15: 183, 1988. J, Woods

Cancer

In: Salmon

VI. Philadelphia: A, Wagner

Limited

E, et al. Twice daily chest irradiation

to etoposide/cisplatin

small cell lung cancer. 106 Turrisi

therapy

W.B. Saunders,

H, Glover

107 McCracken

JD,

Therapy

of

BID thoracic

An ECOG

study

P. ra-

(Abstr.).

230. 1990.

Janaki

LM,

Crowley

for limited

ma: A Southwest

Group

Oncology

carcinoma:

JJ,

et al. Concurrent

small cell lung carcino-

study.

P, Hirsch

irradiation

J Clin Oncol

radiotherapy

including

109 Livingston

8: 892.

A Southwest

brain irradiation Cancer

metastases

scanning

in small cell lung cancer.

bra1 metastases therapy.

RL, Harper

in small

Br J Cancer

124 Lee JS, Murphy

125 Mountain

for extensive

Group

in patients

M. Combined

modality

A Southwest

Treat Rep 70: 1395.

treatment

Oncology

M, Contanzi

of extensive

Group

study.

JL, Bunn PA, Matthews bronchogenic

JJ.

small cell

J Clin Oncol

description,

MJ, et al. CNS metastases

carcinoma:

and changing

pattern

Increasing

in

survival.

Can-

AG, Kristjansen

al irradiation

PEG. Hansen

HH. Prophylactic

and small cell lung cancer.

Cancer

Treat

craniRev 15:

85. 1988.

127 Shepherd

128 Warren

BS, Dhingra

113 Rosen ST, Makuch

RW. Lichter

cranial irradiation

response.

RS, Aisner irradiation

carcinoma. 115 Johnson

Potential

restricted

to patients

remission

and magnetic

with small-cell

E, Klonoff

cancer:

imaging Further

WH. Faber

LP, Gould Cardiovasc

FA.

DH, Einhorn

neuropsychologic,

CJ. McMillan

irradiation

in limited

of

N. The role

stage small cell lung

and CT sequelae.

computed

10 year survivors

of small cell lung cancer.

Int J Radiat

tomography

132 Shepherd

I. Lea

in limited

R, et al. Surgery

small ceil carcinoma

DS, Ruckdeschel

after

initial

of the lung. J

JC, et al. The role of surgery

of selected patients

FA, Ginsberg

A prospective

with small cell carcinoma

irradiation

lung cancer:

and

Dangerous

J Clin Oncol

systemic liaisons?

in 23: 1659,

chemotherapy J Clin Oncol

for 8: 196,

sults of therapeutic

JM, Bunn PA, Glatstein cranial

irradiation

E, Ihde DC. Re-

in small cell lung cancer.

B, Hoskin

of cranial

PJ, Yarnold

JR, Smith IE, Ford

relapse in small cell lung cancer

therapy.

Cancer

Group

study.

133 Salzer GM, Mtiller LC, Huber

and the

Treat Rep 70: 565. 1986.

resection

after chemo-

A University

J Thorac

of Toron-

Cardiovasc

H, et al. Operation

Ann Thorac

Surg 97:

for N2 small

Surg 49: 759, 1990.

JD, Troxell ML, Pazdur

Eyre HJ. Effect of initial resection

R, Coltman

of small-cell

Oncology

carcinoma

Group

of N2 disease in treated small cell carcinoma

CA, of the

Study 7628. J Clin

C, Chretien

cyte levels in bronchogenic gy, clinical

mary

prognostic

effect

of the lung. J Thorac

Surg 88: 495, 1984.

136 Dellon AL, Potvin stage,

PB. Thymus-dependent

carcinoma:

and clinical

Correlations

course

after

lymphowith histolo-

surgical

treatment.

35: 687. 1975. HJ. Rao B, Miyazawa

carcinoma

Thorac

of the lung

Cardiovasc

persensitivity

A, Cohen skin testing

LJ, Wheaton

lor S, Abdou

N. et al. Immune

reactivity

and

to prognosis.

its relation

in priJ

Surg 72: 339, 1976. MH. Fossieck as a prognostic

with small cell lung cancer.

Cancer

indicator

hy-

in patients

52: 1395, 1983.

DL, Manahan-Wahl

N. Lymphocyte

BE, et al. Delayed

subsets

LJ, Sherard

in lung cancer.

S, TayChest 91:

725. 1987. 140 Masuno

Oncol Biol Phys 14: 455, 1988.

120 Lucas CF, Robinson

GA, Evans WK, Feld R.

surgical

for limited small cell lung cancer:

139 Wesselius

1990. J, Crane

RJ, Patterson

study of adjuvant

138 Johnston-Early Brain

of radiation

SD, Greco

tumor

Chest 92: 241, 1987.

for localized

in the management

137 Wanebo

neuropsycho-

scan abnormalities

1985.

HT. Morbidity

I, Williams

of residual

Clin Oncol 5: 1579, 1987.

Cancer

BE, Becker B. Goff WB, et al. Neurologic,

Int J Radiat

neoplasms

Surg 98: 321, 1989.

resection

stage small cell lung cancer.

Cardiovasc

D, Murray

logic, and cranial

119 Carmichael

of the lung.

VE. Neuroendocrine

LH, Mandelbaum

Postchemotherapy

130 Williams

chemotherapy

for small cell carcinoma

135 Meyer JA. Gullo JJ, Ikins PM, et al. Adverse

abnormalifollow-up

Oncol Biol Phys 14: 1109. 1988.

small-cell

Surg 36:

Oncol 3: 964, 1985.

computed

J Clin Oncol 8: 48, 1990. H, Allan B, Graeb

brain

AT.

of the lung. Ann Thorac

lung: A review of Southwest

resonance

lung cancer:

of prophylactic

118 Turrisi

of disease

in stage I and

832, 1988.

134 Friess GG, McCracken

in small cell lung

N, Hayes W, et al. Neurologic,

6- to 13-year survivors.

117 Johnson

PY,

in smallRelation-

SJ, et al. The prospect with chemotherapy

resection

cell lung carcinoma.

Treat Rep 67: 675, 1983.

tomographic,

Clinical,

benefit

J, Wesley MN, et al. Value of prophylactic

Cancer

116 Laukkanen

Holoye

177, 1989.

Am J Med 74: 615, 1983.

given at complete

BE, Patronas

ties in patients

chemo-

272, 1978.

FA, Evans WK, Feld R. et al. Adjuvant surgical

to Lung Oncology

AS, et al. Role of prophylactic

central nervous system metastases

in preventing

in small cell lung cancer: 114 Aroney

HM,

of small cell carcinoma:

of the lung. J Thorac

therapy

with complete

ofcere-

to systemic

of the lung. J Clin Oncol 5: 697, 1987.

cer 44: 1885. 1979. 112 Pedersen

cancer

Semin Oncol5:

combined

131 Baker RR, Ettinger

frequency.

with lengthening

of brain

1017, 1984.

37. 1983.

chemotherapy

cell

Ann

of comput-

for detection

PG, et al. The response

Glisson

therapy.

J Clin Oncol6:

alkylators

2: 585, 1984. I1 1 Nugent

cancer.

61: 147,199O.

WK,

by surgery

following

small cell lung cancer:

study. Cancer

RB, Mira JG, Chen TT. McGavran

lung cancer:

FA. Favorable

J Clin Oncol2:

cell lung

CF. Clinical biology

ship to surgical

129 Johnson

Samson

Greco

cell lung

MJ, Ihde DC, et al. A comparison

and radionuclide

stage II small cell carcinoma

extensive

46: 279, 1980.

S. Mira JG. et al. Combined

irradiation

Oncology

versus

1986. 110 Livingston

impact

ed tomography

control

M. Rygard

small cell anaplastic

of localized

chemotherapy.

RB, Schulman

and multiple-site

trial

prophylactic

combination

FR, Hansen

in bronchogenic

A comparative

receiving

KR,

in small

126 Meyer JA, Comis RL, Ginsberg HH. Dombernowsky

J. Prophylactic

cranial

Hande

Hong WK. Primary chemotherapy of brain metastases cell lung cancer. J Clin Oncol7: 916, 1989.

1990. 108 Hansen

cranial

DH,

metastases

123 Twelves CJ, Souhami

B, Oken M, Bonomi

Concurrent

chemotherapy/radiotherapy

small

of brain

122 Crane JM, Nelson

stage

1990; 162.

D, Mason

with platinum-etoposide:

Proc Am Sot Clin Oncol9:

of limited

S, ed. Adjuvant

stage small cell lung cancer:

diotherapy

L, Johnson

prognosis

Intern Med 106: 386, 1987.

105 Ihde DC, Grayson

and an adjuvant

121 Giannone

T, Ikeda T, Yokota

S. Immunoregulatory small cell lung cancer. 141 Moser

RJ, Tenholder

S, Komuta

T-lymphocyte Cancer MF,

K, Ogura

functions

T, Kishimoto

in patients

with

Res 46: 4195, 1986. Ridenour

ter). Ann Intern Med 103: 478, 1985.

R. Oat-cell

carcinoma

in

132

transfusion

associated

ter). Ann Intern 142 Nusbaum

acquired

immunodeficiency

syndrome

(let-

NJ. Metastatic

small cell carcinoma

of the htng in a pa-

EJ, Orenstein

JM, Chorba

TL, et al. Listeria

genes sepsis and small cell carcinoma presentation

of the acquired

Clin Pathol83: 144 Tsuyuguchi activated

H, Fukuoka

DV, Paschal

M. Increased

BR, Ferree

residue of bacillus

Calmette-Guerin

ma of the lung. Cancer

(MER)

chemotherapy,

carcinoma

Leukemia

B study. J Clin Oncol3:

147 Mikulski

SM, McGuire

Immunotherapy 148 Roszkowski jaszewicz Cancer

Cancer

J, Pulverer

radiotherapy,

small-cell

carcinoma

study. Cancer

PH.

for small-ccl1 46: 2543, 1980. MH,

Chretien

V prolongs patients.

of Human

lung

trials

cancer

in

Cancer.

bined modality

undifferentiated

J

chemoin limited

Oncology

Group

chemoimmuno-

carcinoma

PB, Johnston-Early survival

of the lung.

A, et al. Thymosin

of intensively

treated

small-cell

lung

S, eds. Immunotherapy

New York: Elsevier, B, Chapman

thymosin

of small cell lung cancer.

Cancer

lung cancer: 154 Mabry

First International

Res 48: 1663, of small cell

Lancet

ii: 325, 1987.

antibody

and human

killing of small cell carcinoma

complement

in selective

of the lung. J Clin Invest 75: 1690,

RA, Mabry

cytotoxicity human

M, Sabbath

of murine

small-cell

K, Speak J, Bernal

monoclonal

carcinoma

antibody

in the presence

ment: Possible use for in vitro elimination bone marrow. Int J Cancer 35: 587, 1985. 156 Stahel RA, O’Hara clonal antibody

CJ, Mabry

noma of the lung. Cancer tion of lymphokine-activated against

human

against

of human

comple-

of tumor

cells from

for human

murine

mono-

small cell carci-

killer cell cytotoxicity

by monoclonal

small cell lung carcinoma.

Cancer

sion of class I histocompatibility

Res

antigens,

decreased

protein,

expres-

and mRNA

in human small-ceil lung cancer. J Exp Med 161: 1135, 1985. 159 Ball ED, Sorenson CD, Petengill OS. Expression of myeloid and major

A Phase

histocompatibihty

antigens

on small cell carcinoma

of the

22: 211,

and susceptibility

by natural

to lysis

killer cells. J Immunol HH. Recombinant

of small cell and

squamous

incell

II evaluation

A, Scott E. Small cell lung

of r-interferon-y.

Cancer

60:

2938, 1987. 266 Kristjansen

PEG,

Hansen

HH.

Management

Cancer

of small cell lung

of the Third International

Study of Lung Cancer Workshop

Association

for the

on small cell lung cancer. J Nat1

Inst 82: 263, 1990. K, Niiranen

dose of natural lung cancer:

A, Holsti

a-interferon

L, Pyrhonen

S, Cantell

as maintenance

A Phase III study (Abstr.).

therapy

K. Low-

for small cell

Proc Am Sot Chn Oncol

8: 227, 1989.

169 Valdivieso of

M, Frankmann intravenous

intensive Cancer

BS, Clamon during

GH, et al. Effects of intravenous

treatment

in patients

with small-cell

J Clin Oncol 3: 949, 1985. C, Murphy

WK, et al. Long term ef-

hyperalimentation

chemotherapy

administered

for small cell bronchogenic

during

carcinoma.

59: 362, 1987.

170 Zacharski

LR, Henderson

WG, Rickles FR, et al. Effect of war-

farin on survival in small cell carcinoma ministration

LR, Henderson

farin anticoagulation

Ad-

WG, Rickles FR, et al. Effect of war-

on survival

head and neck, and prostate: Study No. 75. Cancer 172 Chahinian

of the lung: Veterans

study No. 75. J Am Med Assoc 245: 831, 1981. in carcinoma Final

report

of the lung, colon, of VA Cooperative

53: 2046, 1984.

AP, Propert

KJ, Ware JH, et al. A randomized

with

warfarin

and

of alternating

trial chemo-

therapy in extensive small-cell lung cancer by the Cancer Leukemia Group B. J Clin Oncol7: 993, 1989. A, Harvey

HA,

RA233 in the treatment

Walker

B, et al. Chemotherapy

of oat cell lung cancer.

and plus

Am J Clin Oncol

(CCT) 12: 259, 1989. 174 Bronchud

MH, Scarffe JH, Thatcher

of recombinant WJ, Funa K, et al. Markedly

Immunother

AC, et al. Lack of correlation

HFV, Bleehen NM, Galazka

173 Lipton

G, Stone M. Augmenta-

49: 4103, 1989. 158 Doyle A, Martin

therapy

of anticoagulation

Res 46: 2077, 1986.

I57 Tong AW, Lee JC, Wang RM, Ordonez antibodies

LAM2

M, et al. Cytotoxic

LAM8 with specificity

S. Selective

mono-

of the lung: A Phase II study. Eur J Cancer Clin Oncol

carcinoma:

171 Zacharski

1985. 155 Stahel

blood

of tumor cells

23: 987, 1987.

lung cancer.

M, Speak JA, Griffin JD, Stahel RA, Bernal SD. Use of

SM-1 monoclonal

A (IFN-rA)

fraction

LG. Antigens

Workshop.

peripheral

Immunol

MW.

cell lines by

142: 4113, 1989.

fects RL, Beverley PCL, Bobrow

Cancer

of small cellular lung carcinoma

hyperalimentation

1988. 153 Souhami

allogeneic

levels of MHC class I antigen

trial of comV in

Cancer

GD, Fanger tumor

of killing by pretreatment

interferon.

168 Weiner RS, Kramer

1982; 141.

R, et al. Randomized

with and without

of the lung

163 Stam NJ, Kast WM, Voordouw

carcinoma

immuno-

gamma.

1986.

167 Mattson

therapy

the treatment

interferon-activated

cancer: A summary

In: Terry W, Rosenberg

152 Scher HI, Shank

KE, Petter@11 OS, Sorenson

cells: abrogation

and

immunotherapy

versus

T-cells against allogeneic

with interferon

27: 228, 1988.

nuclear

165 Newman

Chemotherapy

treated

gamma

terferon

KP-45.

and induce in small-cell

P, Coulie PG, Van den Eynde B, Symann

of small cell carcinoma

W, Ko H, Jel-

J, et al. Combination

BCG

differentiation

164 Olesen BK, Ernst P, Nissen MH, Hansen

granulosum

of the lung: A Southwest

Cancer Fraction

B, Roszkowski

T, White and

therapy

cancer

F.

49: 2252, 1982.

J, Wiernick

151 Cohen

of clinical

by y-in-

and granulocyte/

inhibit growth

response of human

Immunother

between

109: 72, 1985.

JD, Chen

therapy,

150 Aisner

II. Review

Propionobacterium

Res Clin Oncol

149 McCracken

and

Treat Rev 6: 125, 1979.

G. Small-cell

with

in

M, Muggia

factor

of myeloid

P, Wauters

with gamma

thera-

969, 1985.

WP, Louie AC, Chirigos

K, Nozdryn-Plotnicki

chemotherapy

modality

immunotherapy

of the lung: A Phase III Cancer

of lung cancer:

small cell carcinoma.

Lysis

in small cell carcino-

and

limited small-cell Group

161 Weynants

162 Ball ED, Nichols

chemo-

50: 48,1982.

radiotherapy,

WL, Pert CB. Interferon-y

colony-stimulating

characteristic

Immunol

the methanol-extraction

LH, Pajak T, Eaton W, et al. Combined

py with

Farrar

small cell lung carcinoma

circulating

C, et al. Combined

modulation

lung cancer cell lines. Proc Nat1 Acad Sci USA 83: 6613, 1986.

Am J

Chest 89: 705, 1986.

with and without

by cytofluorography:

Res 46: 2335, 1986.

M, Boon T. Cytolytic

I, Shiratsuchi

therapy-radiotherapy

146 Maurer

An unusual

syndrome.

385, 1985.

T-cells in lung cancer.

145 Jackson

160 Ruff MR, antigens

monocyto-

of the rectum:

immunodeficiency

Cancer

macrophage

tient with AIDS (letter). N Engl J Med 312: 1706, 1985. 143 Read

lung cell lines analyzed terferon.

Med 103: 478, 1985.

patients

receiving

cer. Br J Cancer 175 Bronchud

human

colony-stimulating

N, et al. Phase colony-stimulating

intensive chemotherapy

I/II study factor

in

for small cell lung can-

56: 809, 1987.

MH, Potter

in vivo analysis

granulocyte

MR, Morgenstern

of the effects of recombinant factor in patients.

G, et al. In vitro and human

Br J Cancer

granulocyte

58: 64, 1988.

133

176 Crawford

J. Ozer H, Johnson

cyte colony

stimulating

duced febrile neutropenia A randomized

double

blind

178 0sterhnd

chemotherapy cancer. lung

surviving

without

irradiation:

197 Carmichael

lung

Glutathione

with small cell

chemotherapy

with

Data

on potential

cures, chronic

toxicities,

follow-up.

or

patients

with small

chemotherapy

cell lung

cancer

with or without

treated

irradiation.

survival

of

with combination J Chn Oncol

J, Mitchell

MJ, et al. Non-small

Major cause of late mortality

tients

cell lung

in small cell lung cancer pa-

Am J Med 80: 1103, 1986.

182 Johnson Greco

DH. Porter FA. Acute

nonlymphocytic

small cell lung cancer. 183 0~01s RF, Cowan cancer.

after

treatment

of

Am J Med 81: 962, 1986. and the reversal

Advances

Lippincott,

1986; 129.

in

Oncology

in drug

S, Rosenberg

1986.

The

SA, eds.

Philadelphia:

J.B.

small-cell

DN, et al. Determinants lung cancer

of the sensi-

cell lines to methotrexate.

J Clin Invest 76: 1323, 1985. 185 Curt GA, Carney resistance

DN, Cowan

in human

small-cell

minute chromosomes. 186 Ferro-Luzi

KH, et al. Unstable carcinoma

associated

malian cells: Homology

bacterial

to bacterial

J, Housman

gene: Complete

cDNA

transport

proteins.

protein)

bacterial

Cell 47: 323, 1986.

D. Mammalian

sequence

indicates

in mam-

multidrug strong

resistance

homology

Cell 47: 37 1, 1986.

transport

duplication

gene from multidrug-resistant

and homo-

human

cells. Cell 47: 38 1,

189 Gerlach

JH, Endicott

P-glycoprotein

JA, Juranka

and a bacterial

gests a model for multidrug 190 Batist G, Tulpule KH. Overexpression multidrug-resistant

PF, et al. Homology

haemolysin

resistance.

A, Sinha

Nature

BK, Katki

of a novel anionic human

breast

transport

protein

sug-

324: 485, 1986.

AG, Myers glutathione

cancer

between

transferase

in

cells. J Biol Chem 261:

191 Stevenson

HC, Gazdar

ship between stage small-cell 192 Carmichael

lung cancer.

3, Mitchell

testing of human J Cancer

AF, Linnoila

in vitro tumor

growth

J Chn Oncol7:

JB, DeGraff

lung cancer

RI, et al. Lack of relationand prognosis

in extensive-

923, 1989.

cell lines using the MTT assay.

sion in lung cancer.

LJ, Gottesman J Nat1 Cancer

Br

MM, et al. MDRl gene expresInst 81: Il44,1989.

194 Figueredo A, Arnold A, Goodyear M, et al. Addition of verapamil and tamoxifen to the initial chemotherapy of small cell lung cancer: A Phase I/II study. Cancer

65: 1895, 1990.

lung

Y. GlutaCarcino-

genesis 9: 1681, 1988. H, Tsutsumi

M, Nakajima

tathione&transferase 201 Zijlstra

placental

A, et al. Expression form in human

9: 2325, 1988.

JG, de Vries EGE, Mulder

NH. Multifactorial

sistance in an adriamycin-resistant 202 De Jong DNA

human

JG, De Vries EGE,

topisomerase

II activity

Mulder

NH.

and drug-induced

in an adriamycin-resistant

ma cell line. Cancer

human

small cell lung carcino-

PR. Modifications

of cytotoxic

cyclosporins.

H, Hospers

GAP,

resistance

resistance

cell line. J Nat1 Cancer

in a human

L, Israel MA, et al. Expression

docrine

cell markers

L-DOPA

origin. Cancer

growth

R, Minna

regulation

decarboxylase, tumors

of

small

Inst 81: 1069, 1989.

AF, Helman

in human

by

57: 254, 1988.

Meijer C, et al. Influence

acid on cisplatin

cell lung carcinoma

drug

Br J Cancer

205 Gazdar

endocrine

Reduced

DNA cleavage

Res 50: 304, 1990.

non-immunosuppressive 204 Timmer-Bosscha

drug re-

small cell lung carcino-

Res 47: 1780, 1987.

S, Zijlstra

203 Twentyman

of the glu-

lung carcinomas.

of neuroen-

chromogranin

of endocrine

A,

and non-

Res 48: 4078, 1988.

JD. Opioid

of human

and nicotine

lung cancer

receptors

affect

cell lines. Proc

Nat1

Acad Sci USA 87: 1990. TW, Pert CB, Gazdar

els of intracellular 208 Erisman

bombesin

AF, Carney

D, Minna J. High lev-

characterize

human

small-cell

lung

Science 214: 1246, 1981. MD, Linnoila

zarus LH. Human

RI, Hernandez

lung small-cell

0, DiAugustine

carcinoma

contains

RP, Labombesin.

Proc Nat1 Acad Sci USA 79: 2379, 1982. crine components dysplasias

RI, Remoli

and neoplasms. S, Matsuda

cell carcinoma

VA, Warren

of the bronchopulmonary

WH. Neuroendotract:

Hyperplasias,

Lab Invest 49: 519, 1983.

M, Tateishi

R, Tatsumi

of the lung containing

N, Terazawa

serotonin

T. Oat-

granules.

Gann

59: 123, 1968. 211 Wharton

J, Polak JM, Bloom SR, et al. Bombesin-like in the lung. Nature

212 Yamaguchi

K, Abe K, Kameya

lar size heterogeneity

immuno-

273: 769, 1978. T, et al. Production

of immunoreactive

and molecu-

gastrin-releasing

lung tumors.

Cancer

peptide Res 43:

3932, 1983.

57: 540, 1988.

193 Lai SL, Goldstein

in human

lung tumors.

in fetal and adult lungs and primary

WG, et al. Chemosensitivity

R, Mucilli F, Federici activity

R, Awasthi

reactivity

1986.

transferase

in human

210 Hattori

CE, Cowan

A.

lung cancer

H, Medh

209 Gould VE. Linnoila

1986.

AF, Russo

in human

isoenzymes

carcinoma.

in the mdrl (P-glyco-

proteins

N, Gazdar

activity

AK, Ahmad

207 Moody

to

enzyme

thione-S-transferase

206 Maneckjee

resistance

transport.

188 Chen C, Chin JE, Ueda K, et al. Internal with

with double

Res

9: 335, 1988.

and dense core granules

N Engl J Med 308: 199, 1983.

Ames G. The basis of multidrug

187 Gros P, Croop

methotrexate

Jpn J Cancer

199 Singh SV, Haque

docosahexaenoic

184 Curt GA, Jolivet J, Carney

cell lines correlate

58: 437, 1988.

Carcinogenesis

activity

of resistance

In: DeVita VT, Hellman

Important

tivity of human

leukemia

JD,

K. New aspects of clinical drug resistance:

role of gene amplification refractory

KR. Hainsworth

lung cancer

and carboplatin.

of glutathione

ma cell line. Cancer

LL, List AF, Hande

A

Res 48: 4695,

M, et al. Levels of glutathione-4

JB, Friedman

and related

Carcinogenesis BE, Ihde DC, Matthews

cancer:

logy

to cisplatin

G. Elevation

200 Eimoto

8: 396,

1990. 181 Johnson

Cancer

198 Di Iho C, Del Boccio G, Aceto A, Casaccia tumor.

Ann Intern

RW, et al. Ten-year

patients.

effects

enzymes:

79: 301,1988.

with

combination

J, Makuch

in human

treated

with

BE, Grayson

J, Wada

n mRNA

for small-cell

Med 103: 430, 1985. 180 Johnson

K, Yokota

transferase

cell lines. Br. J Cancer

and late relapses after a five- to eleven-year

drug-metabolizing

study on lung cancer

196 Nakagawa

(Gann)

1044, 1986.

treated

L, et al. Long-lasting

on pulmonary

1988.

P. Mortal-

patients

irradiation

AM, Carrozzi

smoking

with resistance

BE, Ihde DC, Bunn PA, et al. Patients

cancer

(Abstr.).

in lung cancer

M, Dombernowsky

or without

J Clin Oncol4:

179 Johnson

trial

S, Camus

of tobacco case-control

14: 20, 1987.

in long-term

with

controlled

of dose and intensity

HH, Hansen

ity and morbidity

195 Petruzelli

in-

with small cell lung cancer:

placebo

Semin Oncol

K, Hansen

Gramdo-

of chemotherapy

229, 1990.

N. The importance

chemotherapy.

Study Group.

Prevention

in patients

Proc Am Sot Clin Oncol9: 177 Murray

D, G-CSF

factor:

213 Willey JC, Lechner JF, Harris CC. Bombesin and the C-terminal tetradecapeptide of gastrin-releasing peptide are growth factors for normal human bronchial epithehal cells. Exp Cell Res 153: 245, 1984. 214 Cuttitta F, Carney can function cancer.

DN, Mulshine

as autocrine

Nature

growth

316: 823, 1985.

J, et al. Bombesin-like factors in human

peptides

small-cell

lung

134

215 Weber S, Zuckerman fin T. Gastrin lung carcinoma 216 Carney

JE, Bostwick

releasing

F, Moody

peptide.

R, Upp J, Poston

carcinoma modification

bombesin

antagonists

acinar

derivative

with enhanced

antagonist

lung-cancer

binding

inhibits

strong

noma cell line. Biochem 222 Mulshine clonal

antibody

cancer.

to bombesin-like

in murine Swiss 3T3 cells,

subset of human

EA.

recep-

gas&in

J, Avis I, Carrasquillo releasing

peptide

lung cancer (Abstr.). 224 Nakanishi growth

230, 1990.

small cell lung cancer

Kasprzyk

PG,

autocrine

proliferation

cell lines in vitro.

PL, Se&man

small cell lung cancer lular proliferation. in small

cell lung

cholecystokinin, Commun

cancer:

82: 354,

and vasopressin.

neuropeptides Factors

properties

complex

YA, Foglesong is a potent HM,

Becker

channel

an-

hamsters

Correa

of guanine

kinase

oncogenes

several

and tumor suppressor Am J Respir

small-cell

Cell Mel

lung cancer:

Dele-

CS, et al. A non-random

del 3p(14-23)

Genet Cytogenet

in human

small cell

6: 119, 1982.

BE, Minna JD, Sakaguchi

of chromosome

H, Johnson

3p markers

AY. Loss of hete-

in small-cell

B, Hovis J, et al. Molecular 3 in small-cell

BE, Sakaguchi

lung cancer.

analysis

and non-small

AY, Gazdar

of the

cell carci-

AF, et al. Restriction

studies show consistent

3p alleles in small cell lung cancer J, Carritt

at the chromosomal

frag-

loss of chro-

patients’

tumors.

J

B, et al. Deletion

of a DNA sequence

region 3~21 in all major types of lung cancer.

330: 578, 1987.

249 Yokota

AJ. Wad M. Shimosato

of heterozygosity

Y, Terada

on chromosomes

ma and on chromosome

C. J Biol Chem

HP. An animal

Carcinogenesis

model

for

7: 293, 1988.

M, Reznick

neuroendocrine

M, Sugimura

T. Loss

3, 13, I7 in small cell carcino-

3 in adenocarcinoma

of the lung. Proc

GK.

Successful

Cancer

250 Whang-Peng 251 Shiraishi

in

RJH. Evidence

nucleotide-binding

regulatory

by hormones.

Res 50:

for involve-

proteins

FASEB

AG, Watanabe

C phosphorylates

regulatory

the inhibitory

component

function

in hormonal

Biochem

151: 431, 1985.

Y, Bauer S, Jakobs

inhibition

and

in the

J 2: 2569, KH. Pro-

of adenylate

suppresses cyclase.

Eur

S, Noguchi

Jpn J Cancer

retinoblastoma 1988.

Mayo

J

of human

J, Gazdar

in structure

AF, Minna

and expression

in lung cancer.

JM, Park SH, Bogenmann

JD.

of the human

Science 241: 353,

T, Nau MM, Chiba I, et al. ~53: A frequent

tion of the retinoblastoma 255 Takahashi

Y, Sekiya T.

1I in human lung

Res 78: 1302, 1987.

gene in small cell lung cancer.

genetic abnormalities 254 Horowitz

its

M, Shimosato

JW, Lai SL, Whang-Peng

Kaye FJ. Abnormalities

guanine-nucleotide-

apparently

M, Morinaga

carcinomas.

253 Takahashi T, Gilman

in small cell lung carcinoma.

Loss of genes on the short arm of chromosome 252 Harbour

MA, Wojcikiewicz

J. 3p deletion

Clin Proc 64: 256, 1989.

lung tumors

of Ca’+/calmodulin.

of phospholipases

tein kinase binding

Witschi

E. Orloff

with an antagonist

activation 1988. 233 Katada

KL,

of experimental

1645, 1990. 232 Fain JN, Wallace ment

by

small cell lung cancer.

329: 451, 1987.

247 Johnson

Nature

tamoxifen

Res 47: 70, 1987.

RJ, Bell RM. The adriamycin-iron(II1)

lung cancer.

HM,

chemotherapy

of human

Nat1 Acad Sci USA 84: 9252, 1987.

neuroendocrine 231 Schuller

JD, Sausville

transduction

Clin Invest 82: 502, 1988.

264: 9960, 1989. 230 Schuller

Nature

Cancer

248 Kok K, Osinga

antiestrogen

of protein

in a

CS, Lee EC, et al. A specific chromo-

abnormality,

SL, Johnson

rozygosity

mosome

with

bradykinin,

1: 75, 1988.

cell line. Cancer

inhibitor

EA.

mobilization

noma of the lung. N Engl J Med 317: 1109, 1987.

Res

interact

bombesin,

of the antineoplastic

signal

with human

short arm of chromosome

calcium

Biophys

CL, Messing RO. Calcium

in the PC12 neurosecretory 229 Hannun

of cel-

bradykinin.

Biochem

E. Two classes of antagonists

Growth

LM, Sausville

calcium

of lung cancer.

ment length polymorphism

DA, Carpenter

F, Neckers

J, Bunn PA, Kao-Shan

lung cancer.

246 Brauch mobilise

of vasopressin,

and neurotensin.

for the mitogenic

228 Greenberg tagonist

by

164: 66, 1989.

227 Wall PJ, Rozengurt receptors

synthesis

regulator

neuropeptides

Effects

cell lung

Science 215: 181, 1982.

chromosomal

of human

J Clin Invest

cells acts as an autocrine

galanin

small

H, Jensen RT, Minna

inhibits

J, Kao-Shan

244 Whang-Peng

et al. Insulin-like

PA. Transferrin

E. Multiple

Y. Frucht

toxin

tion 3p(1423).

J Clin Invest 82: 33 1, 1988.

226 Wall PJ, Rozengurt

phospholipase-C and

small cell lung cancer cell lines. J Biol Chem 262:

some defect associated

245 Naylor M, Moran

3T3

induce

and the in vitro growth

243 Whang-Peng

with

1988. 225 Vostrejs

JB, Cuttitta

genes in the pathogenesis

I study of anti-

Proc Am Sot Clin Oncol9: JL,

EA. Effect of guanine

Biol 2: 225, 1990. in patients

can mediate

of

in a small-cell

J Clin Invest 86: 1904, 1990.

with lung

antibody

Y, Mulshine factor-I

C

EA. Modulation

hydrolysis

Swiss

242 Viallet J, Minna JD. Dominant

use of a mono-

J, et al. Phase

monoclonal

Cholera

mitogens

156: 1383, 1988.

in patients

kinase

334: 661, 1988.

Res 50: 5257, 1990.

peptides

241 Viallet J. Sharoni

peptide

C.

16456, 1987.

Ann NY Acad Sci 547: 360, 1988.

223 Mulshine

R, Trepel

Bombesin-related

Res Commun

R, Sausville

B, Sausville

from

cells. Cancer

240 Heikkila

AM, et al. Clinical

of protein Nature

on bombesin-stimulated

in membranes

sub-

signals in a small cell lung carci-

Biophys

JL, Avis I, Treston

kinase

cell line. Biochem J 255: 403, 1988.

carcinoma

anti-

F, et al. A novel bombesin

autocrine

of protein

phosphatidylinositol

Y, Viallet J, Trepel

activity

affinities

Proc Nat1 Acad Sci USA 85: 1859, 1988. tor antagonist

binding

and cell signalling.

heterogeneity

JD, Heikkila

and adenine nucleotides

pseudopeptide

small cell lung cancer cells in vitro.

221 Trepel JB, Moyer JD, Cuttitta

perspectives

for cellular regulation.

bombesin-induced

generates

E. [D-Arg’.D-PheS,D-Tryp7,9,Leuri] of human

and

Y. The molecular

239 Sharoni

Swiss 3T3 cells display

bombesin

the growth

Y. Studies

and its implications

J Biol Chem 264: 14691, 1989.

Rozengurt

stance P. a potent inhibits

and

phosphates

238 Trepel JB, Moyer

JE, Jiang NY, et al. Short-chain

activity. PJ,

peptide

MJ, Irvine RF. Inositol

nucleotide

341: 197, 1989.

237 Nishizuka

C, Thomp-

small cell lung

J Biol Chem 264: 11258,1989.

receptor

for pancreatic

releasing

1, 1987.

Science 233: 305, 1986.

V, Townsend of human

by guanine

Mel Cell Endocrinol49:

236 Nishizuka

and

Res 48: 1439, 1988.

of a gastrin

219 Coy DH, Taylor

mitatic

by bombesin

DC, Saari WS, Balishin NL, et al. Carboxyl-terminal

potent antagonists.

220 Woll

growth

on growth

in vivo. Cancer

Nature

Res 47: 821, 1987.

G. Gupta

son J. Effects of bombesin 218 Heimbrook

clonal

Cancer

proteins. 235 Berridge

TW, Minna JD. Selective stimu-

of small cell lung cancer

gastrin-releasing 217 Alexander

for small cell

in vitro. J Clin Invest 75: 306, 1985.

DN, Cuttitta

lation

234 Spiegel AM. Signal transduction

DG, Bensch K, Sikic B, Raf-

peptide is a selective mitogen

target for

Science 246: 491, 1989. E, et al. Frequent

anti-oncogene

is restricted

inactivato a subset

tumor cells. Proc Nat1 Acad Sci USA 87: 2775, 1990. T, D’Amico

D, Chiba

I, Buchhagen

DL. Minna

JD.

135

Identification mechanism

of intronic

point

for p53 inactivation

mutations

as an

in lung cancer.

274 Falco JP. Baylin SB, Lupu R, et al. V-rasH induces

alternative

J Clin Invest 86:

RA, Gerster

JL, Horowitz

acid substitution

results in a retinoblastoma

phosphorylation

and oncoprotein

JM. A single amino protein

binding.

defective

in

Proc Nat1 Acad

Sci

M, Poutska

Homozygous

A, Cavenee

deletion

W, Neve R, Orkin

in Wilm’s tumours

by chromosome

258 Iggo R, Gatter

jumping.

S, Bruns G.

of a zinc-finger

Nature

forms

of p53 oncogene

gene

in primary

expres-

lung cancer.

LF, Weinberg

embryo

oncogenes.

fibroblast

Nature

260 Yancopoulos

requires

L-myc cooperates

two

of

A, Kohl

N, Goldfarb

with rcs to transform

normal

M, cells

JM, Eggleston

primary

rat embryo

fi-

in human

J, Hamilton

S, Baylin S, Vo-

of c-myc and N-myc

in small cell

small cell lung cancer

to chemotherapy

biopsies

expression predicts

and poor prognosis.

amplification

in tumor

patients

of

lack of

Am J Clin Pathol

RW, et al. m_rr family oncogene

cell lines established

and its relationship

from small cell lung

to clinical status

and course.

cancer

BE. Makuch

RW, Simmons

AW. myc family patients’

tumors

DNA

AD, Gazdar

amplification

and corresponding

AF, Burch D.

in small cell lung cell lines. Cancer

Res

A, Nakajima

normality ability.

T. Shiraishi

in lung cancers

Cancer

267 Yokota

and

M, et al. rnyc family gene abits relation

to xenotransplant-

M, Yoshida

cer cells with respect T. Obata

Oncogene

of lung can-

and rearrangement

of

2: 607, 1988.

Y, Sekido Y. et al. Expression

and amplifica-

tion of nryc gene family in small cell lung cancer

and its relation

269 Krystal

characteristics.

Cancer

cancer.

regulation

270 Rodenhuis Zandwijk

S, Van de Wetering pathogenetic

for trans-

in small-cell

lung

ML, Mooi activation

factor

WJ, Evers S, Van

of the K-ras oncogene:

in adenocarcinoma

of the lung. N

S, Slebos RJC, Boot AJM, et al. Incidence

ble clinical significance cinema

of the human

272 Mitsudomi tations

of K-ras oncogene lung. Cancer

T, Viallet J, Linnoila

of rus gene distinguish

Assoc Cancer

activation

and possiin adenocar-

opportunities fostered

R, Minna JD, Gazdar

a subset of non-small

AF. Mu-

cell lung can-

lines (Abstr.).

Proc Am

mones

Lung Cancer

factors

T, Ndkin

cellular

onco-

FASEB

283 Moody

F, Quattrocchi

J 3:

K, Minna

bombesin/GRP-like

J.

peptides

on

Life Sci 37: 105, 1985. M, et al. Effects of peptide

on small cell pulmonary

AF, eds. The Endocrine W.B. Saunders,

KA, Barchas TW,

hor-

carcinoma.

Lung in Health

DN,

is produced

and

1984; 470.

JD. Small cell carcinoma

and receptors. Carney

Neurotensin 284 Moody

Cancer

cell lines contain

57: 769, 1986.

Korman

LY, Gazdar

by and secreted

TW. Neuropeptide

cells. Lung Cancer 285 Cuttitta

A, Minna

J.

from classic small cell

receptors

sertion:

A model for tumor

of human

cancer.

Proc Nat1 Acad Sci USA 85: 6523, 1988.

on small cell lung cancer

4: 186, 1988.

F, Fedorko

J, Gu J, Lebacq-Verheyden

are expressed

in normal

cer: A novel

peptide

human family

AM, Linnoila

peptide gene-associated

peptides

fetal lung and small cell lung canfound

in man.

J Clin Endocrinol

Metab 67: 576. 1988. peptide

F, Vigna SR, et al. Multiple

gene-associated

peptides

are produced

gastrin-releasing by a human

small

cell lung cancer line. J Biol Chem 263: 1928, 1989. 287 Takuwa

N, Takuwa but

neuropeptides

Y, Ohue Y. et al. Stimulation

not

proliferation

in human

CL, Lennon

choline

receptors

by bombesin

small cell lung cancer VA. Activation

inhibits

of calcium and

mo-

tachykinin

cells. Cancer

of M3 muscarinic

voltage-dependent

small cell lung carcinoma.

Res

calcium

acetylinflux

in

J Biol Chem 265: 1443, 1990.

peptide (GRP, mamma289 Viallet J. Minna JD. Gastrin-releasing lian bombesin) in the pathogenesis of lung cancer. Prog Growth Factor

1:89, 1989.

Res

J, Akiyama

retinoblastoma Oncogene

T, Fung Y, et al. Altered

(RB) gene in small-cell

lung cancer. Progress

expression

carcinoma

of the

of the lung.

3: 471, 1988.

MH, Perevodchikova In: Muggia

in Therapeutic

Nl. Single agent F. Rozencweig

Research.

chemotherapy

M, eds. Lung

of

Cancer:

New York: Raven Press. 1979;

343. small-cell 293 Hansen

F, Goldhirsch

lung cancer.

Cancer

SW, Skovsgaard

A. Brunner

KW. New drugs

in

Treat Rev 13: 157, 1986.

T, Sorensen

with 6-mercaptopurine:

JB. Treatment A phase

of small cell

II study.

Cancer

Treat Rep 69: 555, 1985. 294 Murray

N, Shah A, Band P. Phase II study of lonidamine

tients with small cell carcinoma 1283, 1987. 295 Granfortuna

small cell lung

of iproplatin

BD, et al. v-Ha-mm oncogene

progression

involving

4: 176, 1988.

Disease. Philadelphia: 282 Roth

for

G. Rotsch

and growth

in-

Res 32: 289, 1991.

M. Nakagawa

spc-

503, 1988.

by biotechnology.

DN, Cuttitta

receptors

small cell lung cancer.

lung cancer

Res 48: 5738,1988.

cer cell lines from small cell lung cancer 213 Mabry

affinity

292 Joss RA, Cavalli

Engl J Med 3 17: 929, 1987. 271 Rodenhuis

TW, Carney

High

291 Cohen

3373, 1988.

N, Bos J. Mutational

A possible

mechanisms

of the myc gene family

Mol Cell Biol8:

K. DNA sequence

Acta Oncol27:

5, 1989.

290 Yokota

Res 49: 2683, 1989.

G. Birrer M, Way J. et al. Multiple

criptional

BE. Therapeutic

genes: Novel approaches

288 Williams

T. et al. Heterogeneity

to the amplification

myc family oncogenes.

to biological

agents.

50: 240, 1990.

Res 48: 6025. 1988.

J, Wada

268 Takahashi

JA, Lown JW, Mattes WB, Kohn

cificity of antitumor

bilization

48: 5163, 1988. 266 Gemma

277 Hartley

286 Reeve JR. Cuttitta

J Clin Invest 79: 1629. 1987. CashelI

of colorectal

BM. New clue to ras lipid glue. Nature

RI, Battey JF. Gastrin-releasing

BE, lhde DC, Makuch

265 Johnson

cell in a

lung cancer cells. Life Sci 36: 1727, 1985.

L, Nou E, Bergh J. Increased

88: 216. 1987.

cancer

DR, Willumsen

opioid peptides

of the lung. Science 233: 461, 1986.

K, Steinholtz

264 Johnson

non-small

and receptors,

ER, Kern SE, et al. Allelotype

281 Becker KL, Gazdar

J, Kaye F, Sausville EA. Minna JD.

B. Gene amplification

response

factors

Science 244: 207, 1989.

280 Bepler G, Jacques

with TQSto transform

AJ, Ruppert

carcinoma

276 Lowy

human

cooperating

Mol Cell Biol8: 2668, 1988.

gelstein

N-myc

conversion

Proc Nat1 Acad Sci USA 82: 5455. 1985.

broblasts.

263 Funa

at least

GD, Nisen PD. Tesfaye

261 Birrer MJ, Segal S. DeGreve

262 Wong

RA. Tumorigenic

304: 596, 1983.

Ah F. N- ~yc can cooperate in culture.

B, Fearon

carcinomas.

279 Moody

Lancet i: 675, 1990. 259 Land H, Parada

275 Vogelstein

278 Huber

343: 774, 1990.

K, Bartek J, Lane D, Harris AL. Increased

sion of mutant

primary

growth

341: 384. 1989.

USA 87: 6922, 1990.

identified

with associated

small cell lung cancer cell line. J Clin Invest 85: 1740, 1990.

363, 1990. 256 Kaye FJ, Kratzke

257 Gessler

phenotype,

JM, Newman (CHIP)

of the lung. Cancer

N. Ginsberg

in previously

Am J Clin Oncol (CCT)

treated

12: 355, 1989.

in pa-

Treat Rep 7 I :

SJ, et al. Phase II study small-cell

lung cancer.

Small cell carcinoma of the lung: clinical and biologic aspects.

109 Critical Review,s in Oncology/Hematology. 1991; 11: 109-135 0 1991 Elsevier Science Publishers B.V. 1040-8428/91/$3.50 ONCHEM 00006 Small cell...
4MB Sizes 0 Downloads 0 Views