Skip to main content

Full text of "Small cell cancer of the lung : an initial evaluation of the Yale treatment protocol"

See other formats


YALE  MEDICAL  LIBRARY 


3  9002  08676  1872 


PHDTOC0l^ 


YALE 


MEDICAL  LIBRAPY 


z'  / 

Permission  for  photocopying  or  microfilming  of  "  "y  ,, 


for  the  purpose  of  individual  scholarly  consultation  or  refer¬ 
ence  is  hereby  granted  by  the  author.  This  permission  is  not 
to  be  interpreted  as  affecting  publication  of  this  work,  or 
otherwise  placing  it  in  the  public  domain,  and  the  author  re¬ 
serves  all  rights  of  ownership  guaranteed  under  common  law 
protection  of  unpublished  manuscripts. 


(Signature  of  author) 


(Printed  name) 


(Date) 


Digitized  by  the  Internet  Archive 
in  2017  with  funding  from 
Arcadia  Fund 


https://archive.org/detaiis/smaiicellcanceroOObirn 


■i 


SMALL  CELL  CANCER  OF  THE  LUNG: 

AN  INITIAL  EVALUATION  OF  THE  YALE  TREATMENT  PROTOCOL 


A  Thesis  Submitted  to  the  Yale  University 
School  of  Medicine  in  Partial  Fulfillment 
of  the  Requirements  for  the  Degree  of 
Doctor  of  Medicine 


by 


David  Jonathan  Birnkrant 


h\(^c\  1 1  L 


ABSTRACT 


SMALL  CELL  CANCER  OF  THE  LUNG: 

AN  INITIAL  EVALUATION  OF  THE  YALE  TREATMENT  PROTOCOL 
David  Jonathan  Birnkrant 
1985 

A  review  of  small  cell  cancer  of  the  lung  (SCCL)  is  presented, 
followed  by  initial  results  from  the  Yale  University  treatment 
protocol  for  SCCL. 

The  review  includes  selected  topics  on  the  epidemiology, 
etiology,  cytogenetics,  cytomorphology ,  cells  of  origin,  cell 
products,  clinical  diagnosis,  staging,  prognosis,  and  treatment  of 
SCCL.  The  tumor  emerges  as  one  of  diverse  clinical  behavior  and 
cellular  character;  it  remains  poorly  understood.  Despite 
intermediate  or  slow  doubling  time,  dissemination  of  the 
tumor — microscopic  or  gross — is  the  rule  at  diagnosis.  Modest  success 
has  been  achieved  in  treatment,  allowing  a  small  group  of  patients  to 
go  on  to  long-term  survival  (4  or  5  years),  but  most  patients  relapse 
after  initial  response  to  therapy  and  second-line  therapy  is  rarely 
effective.  Special  attention  is  paid  to  treatment  design,  including 
the  role  of  prophylactic  cranial  irradiation  (PCI),  adjuvant  radiation 
therapy,  and  the  concept  of  local  tumor  control. 

Results  from  the  Yale  University  treatment  protocol  are 
presented.  T-hirty-nine  evaluable  patients  were  prospectively 
randomized  to  therapy  with  cyclophosphamide,  Adriamycin,  and 
vincristine  (CAV)  every  21  days  or  CAV  alternating  with  Etoposide 


(VP-16-213).  All  limited  disease  (LD)  patients  underwent  thoracic 
irradiation;  complete  responders  received  PCI.  Eighty  percent  of  LD 
patients  achieved  complete  response  as  did  12%  of  extensive  disease 
(ED)  patients.  Projected  median  survival  ranged  from  198  days  for  ED 
non-responders  to  560  days  for  LD  complete  responders.  It  is  too 
early  to  report  on  long-term  survival.  Addition  of  Etoposide  (E) 
afforded  no  significant  survival  advantage  over  the  CAV  regimen. 
Etoposide  added  no  significant  additional  toxicity;  in  fact,  the 
percentage  of  patients  experiencing  infections  requiring 
hospitalization  was  lower  in  the  CAV/E  group  (6%  vs.  33%).  This  may 
be  the  result  of  a  smaller  proportion  of  patients  on  CAV/E 
experiencing  leukopenia  (44%  vs.  76%). 


ACKNOWLEDGEMENTS 


Dr.  Carol  Portlock  was  the  perfect  thesis  advisor  (_g^<.0001).  Her 
scholarship  was  complemented  by  a  commitment  to  scientific 
open-mindedness — which  is  to  say:  She  came  to  the  rescue  when  I  was 
up  to  my  elbows  in  it,  but  never  imposed  her  point  of  view.  Thus  I 
had  the  happy  experience  of  learning  through  discovery.  Her  gracious 
good  humor  was  infectious  and  her  tolerance  of  my  eccentric  work 
habits  wonderful.  Thanks  to  her,  this  thesis  was  transformed  from  a 
requirement  for  graduation  into  a  rewarding  scientific  project. 

Dr.  Diana  Fischer  was  my  Virgil  through  the  inferno  of 
statistics.  Because  she  never  discussed  this  study  except  in  the 
broader  context  of  statistical  concepts,  I’ve  gained  a  sense  of  the 
fundamentals  of  experimental  design.  Dr.  Fischer  shared  her  time, 
knowledge,  and  enthusiasm  with  a  generosity  for  which  I  am  very 
grateful . 

Mrs.  Terry  O'Connor  did  the  most  extraordinary  things  with  a 
computer  and  she  did  them  better  and  faster  than  I  had  thought 
possible.  No  deadline  was  so  pressing  as  to  disturb  her  reassuring 
calm.  I  am  lucky  to  have  enjoyed  the  benefit  of  her  expertise  and 
good-naturedness. 

Mrs.  Rosemary  Slattery  was  always  ready  with  all  manner  of 
encouraging  words  and  warm  smiles.  When  things  got  rough,  she  would 
regale  me  with  her  classic  answer  to  my  desperate  "Rosemary,  will  Dr. 
Portlock  be  around  next  week?":  "Well,  I've  worked  with  her  for  eight 
years,  and  she  hasn't  taken  a  vacation  yet...." 


r 


A  large  "thank  you"  to  the  physicians  who  made  me  welcome  in 
their  offices,  made  it  possible  for  me  to  review  patients  charts,  and 
made  time  to  answer  my  questions:  Drs.  Fischer  and  Wiggans;  Dr.  M.E. 
Katz;  Drs.  Levy,  Farber,  Bobrow,  and  Lundberg;  Dr.  J.E.  Brown;  Dr. 

G.T.  Kenneally;  and  Dr.  I.S.  Lowenthal. 

Dr.  J.  Bernard  Gee  read  this  thesis  for  the  Department  of 
Internal  Medicine.  His  criticisms  and  comments  were  uniformly 
helpful.  At  his  suggestion,  I  was  in  contact  with... 

Dr.  Raymond  Yesner,  who  very  kindly  took  the  time  to  meet  with  me 
on  an  "emergency"  basis.  Dr.  Yesner ’s  summary  of  the  conclusions 
reached  at  the  September,  1984  meeting  of  the  pathology  panel  of  the 
International  Association  for  the  Study  of  Lung  Cancer  made  it 
possible  for  me  to  include  mention  of  an  exciting  new  approach  to  the 
histological  classification  of  small  cell  lung  cancer  in  this  paper. 

Thank  you,  Mrs.  Fran  DeGrenier,  for  typing  strange  words  at 
strange  hours  and  meeting  every  deadline. 

Finally,  if  a  dedication  were  appropriate  for  so  modest  a  piece 
of  work,  it  would  be  to  my  parents,  with  love. 


TABLE  OF  CONTENTS 


PART  I:  THE  BASICS  1 

Introduction  1 

Epidemiology  and  Etiology  2 

Cytogenetics,  Cytomorphology ,  Cells  of  Origin  and 

Cell  Products  5 

Clinical  Diagnosis  16 

Staging  and  Prognosis  21 

PART  II:  TREATMENT  30 

Overview  30 

Treatment  of  Limited-Stage  Disease  and  Treatment- 

Related  Toxicities  36 

Extensive-Stage  Disease  and  Experimental  Therapies  45 

Closing  Comments  49 

PART  III:  THE  STUDY  53 

Methods  53 

Patients  and  Results  57 

Discussion  67 

REFERENCES  71 

TABLES 

Table  1:  Age  Standardized  Percentage  Distribution  of 

SCCL  by  Cigarette  Smoking  Habit  85 

Table  2:  Distribution  of  Excess  (Presumably  Radiation- 

Induced)  Bronchogenic  Cancers  by  Radiation 
Exposure  Group  86 

Table  3:  Symptoms  of  SCCL  87 

Table  4:  Percent  Distribution  of  Metastases  at 

Presentation  and  at  Autopsy  in  Two 
Studies  88 

Table  5:  Recommendations  for  Restaging  89 

Table  6:  Prognostic  Factors  in  SCCL 


90 


I 


Table 

7: 

Treatment  Results  in  SCCL:  Selected  Studies 

91 

Table 

8; 

Long-Term  Survival  (>_5  years)  in  SCCL 

92 

Table 

9: 

Treatment  Protocol  for  Limited  Disease 

93 

Table 

10: 

Treatment  Protocol  for  Extensive  Disease 

94 

Table 

11: 

Patient  Characteristics  by  Treatment  Arm 

95 

Table 

12: 

Metastatic  Sites  at  Diagnosis  in  Extensive 
Disease  (ED)  Patients  by  Treatment  Arm 

96 

Table 

13: 

Sites  of  Relapse 

97 

Table 

14: 

Treatment  Results:  Response  and  Time  to 
Relapse 

98 

Table 

15: 

Survival 

99 

FIGURES 


Figure 

1: 

Time  to  Relapse  for  Responders  (CR  +  PR) 
Disease  Extent 

by 

100 

Figure 

2: 

Time  to  Relapse  for  Extensive  Disease 
Patients  by  Response  Group 

101 

Figure 

3: 

Time  to  Relapse  for  Limited  Disease 
Patients  by  Treatment  Group 

102 

Figure 

4: 

Time  to  Relapse  for  Extensive  Disease 
Patients  by  Treatment  Group 

103 

Figure 

5: 

Overall  Survival 

104 

Figure 

6: 

Survival  for  Responders  (CR  +  PR)  by 
Disease  Extent 

105 

Figure 

7: 

Survival  for  Extensive  Disease  Patients 
Response  Group 

by 

106 

Figure 

8: 

Survival  for  Limited  Disease  Patients  by 
Treatment  Group 

107 

Figure 

9: 

Survival  for  Extensive  Disease  Patients 
Treatment  Group 

by 

108 

List  of  Abbreviations 


A 

ACTH 

ADH 

APUD 

BN 

C 

CBC 

CEA 

CK-BB 

CNS 

CR 

CT 

DDC 

DNA 

E 

ECOG 

ED 

EKG 

HCG 

INH 

LD 

LDH 

LI 

M 

MoAb 

NCI 

NR 

NSE 

P 

PCI 

PPD 

PR 

PS 

PTH 

RT 

SIADH 

SVC 

TNM 

V 

VA 

WHO 


Adriamycin  (doxorubicin) 
adrenocorticotropic  hormone 
antidiuretic  hormone 

Amine  Precursor  Uptake  and  Decarboxylation 
bombesin 

cyclophosphamide 
complete  blood  count 
carcinoembryonic  antigen 
BB  isoenzyme  of  creatine  kinase 
central  nervous  system 
complete  response 
computerized  tomography 
dopa  decarboxylase 
deoxyribonucleic  acid 
Etoposide  (VP-16-213) 

Eastern  Cooperative  Oncology  Group 

extensive-stage  disease 

electrocardiogram 

human  chorionic  gonadotropin 

isoniazid 

limited-stage  disease 
lactic  dehydrogenase 
labeling  index 
methotrexate 
monoclonal  antibody 
National  Cancer  Institute 
non-response 
neuron  specific  enolase 
cis-platin 

prophylactic  cranial  irradiation 
purified  protein  derivative 
partial  response 
performance  status 
parathyroid  hormone 
radiation  therapy 

syndrome  of  inappropriate  secretion  of  antidiuretic  hormone 
superior  vena  cava 
tumor -nodes-me t as tases 
vincristine 

Veterans  Administration 
World  Health  Organization 


1 


SMALL  CELL  CANCER  OF  THE  LUNG: 

AN  INITIAL  EVALUATION  OF  THE  YALE  TREATMENT  PROTOCOL 
PART  ONE:  THE  BASICS 
Introduction 

Small  cell  cancer  of  the  lung  (SCCL)  is  a  disease  whose 
significant  incidence  and  poor  prognosis  make  it  of  major  concern. 

This  is  especially  true  in  a  society  like  ours,  with  its  increasing 
emphasis  on  preventive  medicine  and  the  containment  of  health  care 
costs.  The  etiologic  factors  in  the  disease — tobacco  smoking, 
ionizing  radiation,  asbestos  and  chemicals — make  SCCL  somewhat 
preventable.  Treatment  for  SCCL  is  often  initially  effective,  but 
relapse  is  the  rule  and  the  disease  then  proves  resistant  to  second- 
line  therapies.  In  light  of  the  relatively  poor  results  achieved  with 
today’s  state-of-the-art  therapy,  it  would  be  wise  to  direct  concerted 
efforts  toward  prevention  of  SCCL.^  Unfortunately,  the  medical 
profession  has  made  no  successful  attempt  to  wrest  responsibility  for 
such  a  goal  from  businessmen  and  politicians.  As  long  as  tobacco 
remains  an  important,  heavily  subsidized  cash  crop  and  images  from 
Madison  Avenue  dictate  our  behavior,  the  individual  medical 
practitioner-haranguing  his  patients  about  smoking — will  remain  at 
best  a  nagger,  at  worst  a  bore. 

This  paper  will  emphasize  the  diversity  in  data  on  SCCL.  Even 
the  most  basic  aspects  of  the  disease  are  poorly  understood  and  the 
lack  of  an  effective  treatment  strategy  reflects  that  ignorance.  The 


first  part  of  the  paper  takes  the  form  of  a  review;  the  second  part 
presents  initial  data  from  the  Yale  University  treatment  protocol  for 
SCCL. 


2 


Epidemiology  and  Etiology 

Lung  cancer  death  rates  have  been  increasing  at  a  spectacular 

2 

rate  when  compared  to  that  of  other  cancers.  After  World  War  II, 

lung  cancer  death  rates  for  men  began  rising  at  a  faster  rate  than 

straightline  projections  would  have  predicted.  The  rate  curve  for 

1  2 

women  has  followed  suit  for  the  last  twenty  years.  ’  Projections  for 

1984  show  that  lung  cancer  will  be  the  most  common  cause  of  cancer 

deaths  for  men  and  the  second  most  common  cause  of  cancer  deaths  for 
2 

women . 

3 

Weiss  has  reviewed  numerous  studies  on  the  incidence  of  SCCL. 

He  reports  W.E.  Morton's  unpublished  data  on  SCCL  rates  in  Portland, 

Oregon  for  the  period  1968-1972.  The  disease  was  more  frequent  in  men 

than  women.  Mean  annual  rates  per  100,000  population  were  13  for 

4 

males,  4  for  females.  In  contrast,  Annegers  et  al.  reported  a  rate 
of  6/100,000  among  males  in  rural  Olmstead  County,  Minnesota  for  the 
period  1965-1974.  This  lower  rate  is  consistent  with  the  notion  that 
SCCL  is  less  common  in  rural  areas. ^ 

The  median  age  at  diagnosis  in  most  series  is  about  60  years. ^ 

The  Philadelphia  Pulmonary  Research  Project  studied  the  natural 
history  of  lung  cancer  in  men  45  years  of  age  and  older,  each  of  whom 
was  followed  for  ten  years.  Unpublished  data  from  the  study,  quoted 


3 


3 

by  Weiss,  show  the  highest  incidence  of  disease  among  men  55-64  years 

of  age.  The  study  reported  the  highest  rate  of  lung  cancers  in 

general,  irrespective  of  subtype,  in  the  60-  to  64-year-old  age 

4 

group.  In  contrast,  Annegers  et  al.  found  their  highest  incidence 

rate  among  men  75  years  or  older  for  the  period  1965-1974. 

SCCL  is  generally  quoted  as  accounting  for  20-25%  of  all  lung 

cancers.^  When  the  data  are  examined,  however,  the  frequency  of  small 

cell  as  a  percentage  of  all  typed  cancers  varies  widely.  In  Weiss's 
3 

review,  SCCL  accounts  for  13.7%  -  39.6%  of  all  typed  lung  cancers 

among  men  and  9.4%  -  31.3%  of  lung  cancers  among  women,  depending  on 

the  study  consulted.  Perhaps  these  ranges  represent  differences  in 

histological  interpretation,  or  differences  in  incidence  related  to 

the  population  observed  (the  highest  relative  incidence  figures  come 

8  9 

from  a  study  done  in  Iceland  ).  Kyriakos  and  Webber,  reporting  on 
lung  cancer  in  young  adults,  have  found  a  rate  of  13%  SCCL  among 
patients  of  all  ages  with  lung  cancer  at  Barnes  Hospital,  St.  Louis. 
Their  review  of  the  literature  revealed  that  SCCL  accounted  for  2-38% 
of  lung  cancers  in  several  large  series.  Interestingly,  these  authors 
found  a  slightly  higher  relative  incidence  of  SCCL  (24%)  among  younger 
patients  (45  years  of  age  or  less).  Kennedy^^  found  a  remarkably  high 
proportion  of  small  cell  tumors  (65%)  in  his  series  of  40  lung  cancers 
occurring  in  patients  under  the  age  of  40  in  England.  In  contrast, 
Putnam^ ^  at  Walter  Reed  Army  Medical  Center  found  17%  small  cell 
tumors  among  24  patients  with  lung  cancer  under  the  age  of  40. 


4 


The  male: female  ratio  of  SCCL  is  generally  thought  to  be  higher 

than  for  other  types  of  lung  cancer,  with  a  ratio  of  3  males:!  female 

3 

reported  by  Morton. 

Etiological  factors  in  SCCL  include  exposure  to  tobacco  smoking, 

ionizing  radiation,  asbestos,  chemicals,  metals,  and  possibly  air 

pollution.  Pathogenesis  of  the  disease  is  not  well  understood;  the 

tumors  are  usually  central  in  location  and  are  presumed  to  arise,  like 

squamous  cell  carcinoma,  through  chronic  irritation,  mucosal 

12 

denudement,  and  absorption  of  carcinogens.  The  cells  of  origin  of 

the  tumor  are  of  great  interest  and  will  be  discussed  later  on. 

There  is  probably  a  dose-response  relationship  between  cigarette 

consumption  and  the  development  of  SCCL.  The  Philadelphia  Pulmonary 

13 

Neoplasm  Research  Project's  data  supported  this  notion.  Auerbach  et 
14 

al.  found  that  small  cell  as  a  percentage  of  all  lung  cancers  rose 

from  14.5%  for  ex-smokers  progressively  to  31.1%  for  those  patients 

smoking  more  than  two  packs  a  day.  (See  Table  1.)  This  rise  was  not 

seen  in  other  histological  subtypes.  Of  note  is  the  possibility  that 

cessation  of  smoking  is  associated  with  longer  survival  in  SCCL,  even 

when  patients  stop  at  the  time  of  diagnosis. 

The  link  between  exposure  to  ionizing  radiation  and  SCCL  is  a 

1 6 

strong  one.  Archer  et  al.  compared  rates  of  lung  cancer  and  its 
subtypes  among  uranium  miners  to  the  rates  expected  for  a  matched 
group  without  radiation  exposure.  The  authors  expected  to  find  14.06 
respiratory  cancers  in  their  study  group.  Instead,  107  cases  were 
recorded  among  the  miners,  66  of  which  were  SCCL  (62%).  In  the 


5 


matched  control  group,  SCCL  should  have  accounted  for  14.05%  of  the 

lung  cancers.  There  was,  in  addition,  a  dose-response  relationship 

found  for  SCCL  with  increasing  radiation  exposure.  These  data  appear 

in  Table  2.  Unlike  smoking,  then,  radiation  exposure  may  produce  a 

predominance  of  small  cell  cancers  in  the  lung.  Agreement  on  this 

point  is  not  universal,  however.^ 

Asbestos  and  chemicals  have  been  implicated  in  the  etiology  of 

lung  cancers  in  general.  Although  data  on  subtypes  is  limited,  SCCL 

appears  to  be  associated  with  exposure  to  these  agents.  The  role  of 

smoking  in  the  carcinogenicity  of  asbestos  is  still  not  clear;  is 

asbestos  a  carcinogen  or  a  synergistic  agent  which  promotes  cancer  in 

smokers?  The  chemicals  for  which  there  is  evidence  of  carcinogenicity 

in  humans  include:  "polycyclic  aromatic  hydrocarbons,  certain  metals 

3 

or  their  compounds,  and  certain  simple  organic  chemicals."  The  list 
of  possible  carcinogens  grows  longer  every  day,  but  the  strict 
scientific  criteria  needed  to  show  causality  make  such  proof  a  task 
that  is  pain-staking,  if  not  impossible. 

Cytogenetics,  Cytomorphology ,  Cells  of  Origin, 
and  Cell  Products 

The  basis  of  understanding  the  small  cell  tumor  lies  in  an 
understanding  of  its  component  cells.  One  approach  to  these  cells  is 
through  cytogenetic  studies.  Do  the  chromosomes  of  the  tumor  have 
identifiable  characteristics? 


6 


Wurster-Hill  and  Maurer  studied  the  chromosomes  of  patients' 

SCCL  tumors  using  direct  bone  marrow  preparations  and  trypsin-Giemsa 

banding.  Chromosome  number  and  structural  aberrations  (markers)  were 

frequent  and  highly  variable.  Chromosome  number  (ploidy,  DNA  index) 

in  untreated  patients  ranged  from  hypodiploid  to  polyploid  with  the 

latter  most  common  (the  chromosome  count  was  typically  in  the 

80's).  A  structural  abnormality  of  chromosome  #1  was  found  in  14  of 

the  18  patients  with  karyotypic  abnormalities  (total  patients  =  26). 

But  very  few  markers  were  common  to  two  or  more  patients  and  the 

consistency  of  given  markers  among  the  cells  from  one  patient  was 

usually  poor.  The  presence  of  cells  with  different  abnormalities  of 

chromosome  number  in  the  same  patient  (e.g.,  diploid  and  polyploid) 

18 

was  discovered.  Vindelov  et  al.  found  that  ploidy  in  their  SCCL 

tumor  cells  could  be  grouped  into  near-diploid,  near-triploid ,  and 

near-tetraploid  values.  Each  of  five  patients  was  found  to  have  two 

clones  with  different  chromosome  number  in  a  single  metastasis  (17%  of 

the  total  patients).  The  authors  view  this  as  evidence  that  SCCL,  at 

least  for  some  patients,  is  not  monoclonal.  That  is:  new  cell  lines 

evolve  from  the  original  tumor.  These  cell  lines  may  have 

characteristics  (clinical,  biochemical)  that  are  entirely  different 

from  those  of  the  original  tumor.  The  heterogeneity  of  the  SCCL 

tumor — a  concept  emphasized  in  this  paper — may  lie  in  the  evolution  of 

more  than  one  cell  line  from  the  original  tumor. 

19 

Whang-Peng  et  al.  found  two  distinct  stem  lines  in  2  of  their 
12  cell  lines  cultured  from  human  small  cell  lung  cancer  tissue. 


7 


These  authors  describe  a  consistent,  acquired  chromosomal 
abnormality — a  deletion  in  the  short  arm  of  chromosome  #3 — present 
both  in  SCCL  cell  lines  and  in  fresh  clinical  specimens  cultured  for  2 
days  in  a  serum-free  medium.  Chromosome  studies  of  other  types  of 
neoplasms  have  not  shown  a  specific  abnormality  of  chromosome  #3.  The 
data  of  Wurster-Hill  and  Maurer  (abnormality  of  chromosome  #1)  do  not 
fit  easily  into  the  scheme  of  Whang-Peng  et  al.;  an  explanation  of  the 
discrepancies  awaits  elucidation.  Whang-Peng  et  al .  appear  to  have 
found  a  specific,  acquired  somatic  cell  defect  (deletion  3p,  14-23) 
associated  with  continued  replication  of  SCCL  tumor  cells.  If  this 
holds  true,  the  diagnosis  and  treatment  of  SCCL  will  be 
aided — especially  if  the  function  of  the  genes  present  in  the  region 
of  chromosome  #3  where  the  deletion  was  found  can  be  understood. 

Another  approach  to  the  cells  of  the  tumor  is  to  ask:  where  does 
SCCL  arise  from?  That  is,  which  cells  in  the  lung  first  acquire  the 
chromosomal  abnormalities,  due  to  exposure  to  carcinogens,  which  lead 

to  the  growth  of  a  tumor? 

20 

Hattori  et  al.  studied  24  cases  of  oat-cell  (small  cell) 
carcinoma  of  the  lung  and  four  cases  of  bronchial  carcinoid  tumor  both 
under  the  electron  microscope  and  biochemically.  They  found  that  SCCL 
tumor  cells  were  characterized  by  the  presence  of  neurosecretory-type 
granules  (NSGs)  of  800-2000  Angstroms,  almost  identical  to  but 
somewhat  smaller  than  the  NSGs  found  in  4  samples  of  bronchial 
carcinoid  tumor .  NSGs  were  not  found  in  139  samples  of  other  types  of 
lung  tumors  studied.  Serum  serotonin  level  was  elevated  in  13  of  20 


8 


small  cell  cases  and  the  degree  of  elevation  seemed  to  correlate  with 

the  number  of  NSGs  present  in  the  tumor  cells.  Serotonin  level  in 

tumor  tissue  was  elevated  in  7  of  12  cases  of  SCCL  but  in  only  1  of  4 

cases  of  bronchial  carcinoid  tumor.  In  5  of  7  cases  of  SCCL,  both 

serotonin  and  ACTH  were  elevated  in  tumor  tissue  samples.  Other  types 

of  lung  cancer,  which  lacked  NSGs,  showed  no  elevation  of  serotonin 

activity  with  the  exception  of  one  case  of  squamous  cell  carcinoma 

(and  one  case  of  pleurisy  due  to  collagen  vascular  disease).  The 

authors  noted  that  the  NSGs  they  found  in  bronchial  carcinoid  and  SCCL 

were  identical  to  those  which  had  been  previously  described  by  Bensch 
21 

et  al .  in  the  Kulchitsky-type  cells  in  bronchial  mucus  glands. 

Hattori  et  al.  thus  concluded  that  "oat-cell  carcinoma  is  a  special 

type  of  lung  tumor  producing  neurosecretory-type  granules  and  a  highly 

malignant  variant  of  bronchial  carcinoid  tumor  which  is  originated 

from  neurosecretory-type  cell  (Kulchitsky-type  cell)  found  in 

20 

bronchial  glands."  This  is  a  remarkable  statement,  for  the  authors 

have  found  an  association  between  electron  microscopic  characteristics 

of  the  tumor  (NSGs)  and  tumor  products  (serotonin,  ACTH)  known  to  have 

22 

clinical  significance  as  ectopic  hormone  products  of  SCCL  tumors. 

Moreover,  the  ultrastructure  of  the  tumor  cells  has  provided  a  clue  to 

cell  origin:  the  Kulchitsky-type  cells  of  bronchial  glands.  The 

latter  can  produce  serotonin  from  tryptophan  and  5-hydroxy-tryptophan, 

which  means  they  fit  into  Pearse’s  conception  of  an  APUD  (Amine 

23 

Precursor  Uptake  and  Decarboxylation)  cell.  APUD  origin  would,  in 
turn,  imply  a  way  of  attacking  the  embryological  lineage  of  SCCL ' s 


9 


cells  of  origin  and  relate  SCCL  to  other  cells  of  APUD  origin  in  the 

body.  In  fact,  the  above  associations  reach  beyond  our  ability  to 

24 

apply  them.  Indeed,  Hattori  et  al.  examined  the  cytomorphology  of 

SCCL  tumors  in  relation  to  response  to  therapy  in  an  article  published 

five  years  after  the  one  already  discussed.  Surprisingly,  although 

almost  all  SCCL  tumor  cells  were  found  to  contain  NSGs,  the  cells  from 

tumors  which  did  not  respond  to  combination  chemotherapy  showed  few  or 

no  NSGs.  Some  of  the  tissue  specimens  used  in  the  study  were  obtained 

from  autopsy  material,  and  it  is  possible  that  chemotherapy  affected 

the  cell  structure.  Alternatively,  a  cell  line  without  NSGs  could 

have  arisen  from  the  original  tumor.  Still,  the  lack  of  NSGs  in  the 

non-responder  group  shakes  the  foundation  of  attempts  to  characterize 

SCCL  at  an  ultrastructural  level  in  a  way  that  is  consistent  with  the 

presumed  cells  of  origin. 

25 

Tischler  notes  that  the  APUD  concept  has  been  elucidated  and 

revised  since  its  initial  introduction.  APUD  cells  are  now  known  to 

occur  in  two  distributions  in  the  lung:  as  scattered  Kulchitsky-like 

cells  and  as  organized  groups  of  cells  called  neuroepithelial  bodies. 

Both  occur  in  close  proximity  to  nerve  endings.  The  secretory 

products  and  physiological  role  of  APUD  cells  in  the  lung  are 

obscure.  While  the  APUD  concept  may  explain  the  source  of  some  of  the 

hormones  SCCL  tumors  produce,  cytogenetic  abnormalities — "derepression 

of  the  genome" — might  also  account  for  production  of  ectopic 

26 


hormones. 


10 


Still,  APUD  has  been  used  as  a  window  to  the  study  of  SCCL’s 

ectopic  hormones.  A  wide  variety  of  such  hormones  have  been 

identified  and  include  ACTH,  ADH,  calcitonin,  glucagon,  HCG, 

18  22  27 

serotonin,  PTH,  and  estradiol.  ’  ’  The  frequency  of  ectopic 

hormone  production  by  small  cell  tumors  is  not  known;  it  is  clear, 

however,  that  clinically  apparent  syndromes  that  can  be  traced  to 

these  hormones  are  much  rarer  than  production  of  the  hormones 

themselves.  Thus,  while  more  than  50%  of  patients  may  have  abnormally 

high  levels  of  hormones  such  as  ACTH,  ADH  and  calcitonin,  clinical 

syndromes  such  as  SIADH  or  Cushing's  syndrome  appear  to  occur  in  less 

28 

than  10%  of  patients.  Richardson  et  al .  point  out  that  the  identity 
of  these  tumor-produced  "ectopic  hormones"  is  not  really  known;  they 
may  or  may  not  be  identical  to  "normal"  hormones  and  radioimmunoassays 

r  28 

of  these  substances  may  be  neither  truly  sensitive  nor  specific. 

29 

Despite  these  caveats.  Science  marches  on.  Baylin  and  Gazdar 
have  measured  biochemical  indices  in  SCCL  with  established 
relationships  to  APUD  cells  outside  the  lung.  These  include  L-dopa 
decarboxylase  (central  to  the  APUD  concept — it  converts  precursor 
amino  acids  into  their  corresponding  amines);  and  calcitonin  (produced 
by  the  APUD  tumor  medullary  thyroid  carcinoma).  They  also  measured 
histaminase  and  beta-endorphin,  neither  of  which  is  specific  to  APUD 
cell  activity;  still,  both  substances  are  thought  to  be  involved  in 
hormone  production  by  cancers.  The  authors  found  that  the  biochemical 
parameters  studied  were  not  specific  to  SCCL  and  that  there  was  great 
heterogeneity  of  findings  between  different  patients  with  SCCL,  among 


11 


different  lesion  sites  (primary  vs.  metastatic)  in  the  same  patient 
with  SCCL,  and  even  within  individual  SCCL  lesions.  These  data  are 
viewed  as  a  reflection  of  heterogeneous  cell  populations  all  of  which 
are  grouped  together  under  the  clinical  term  "small  cell  lung 
cancer."  The  authors  noted  that  quantitatively,  however,  their 
endocrine  parameters  tended  to  group  more  with  SCCL  than  with  other 
lung  tumors. 

An  attempt  has  been  made  to  find  tumor  products  which  will  prove 
to  be  specific  markers  of  SCCL,  whose  concentrations  are  proportional 
to  tumor  burden,  and  which  are  present  in  enough  patients  to  make 
measurement  worthwhile.  Three  such  newly  described  products  are 
bombesin,  the  BB  isoenzyme  of  creatine  kinase  (CK-BB),  and 
neuron-specific  enolase  (NSE).  I  will  describe  a  study  of  NSE  as  an 
example . 

30 

Carney  et  al .  studied  serum  NSE  levels  at  diagnosis  in  94 
patients  with  SCCL.  The  levels  were  repeated  during  and  after 
therapy.  Sixty-nine  percent  of  all  patients  had  a  serum  NSE  level 
more  than  3  S.D.  above  control,  including  87%  of  the  patients  with 
extensive-stage  disease  (ED).  Mean  serum  NSE  was  significantly  higher 
in  patients  with  ED  than  in  those  with  limited-stage  disease.  In  20 
of  21  patients,  all  of  whom  had  elevated  NSE  levels  at  diagnosis, 
serum  NSE  fell  significantly  as  the  patients  responded  to 
chemotherapy.  In  the  one  patient  whose  level  remained  unchanged,  no 
response  to  therapy  was  achieved  and  the  disease  progressed.  The  NSE 
level  dropped  into  the  normal  range  for  all  patients  achieving  a 


12 


complete  response. 

Serial  NSE  measurements  showed  a  good  correlation  between 

clinical  condition  and  the  level  of  the  marker.  For  example,  in  9 

patients  with  raised  NSE  levels  on  diagnosis  the  levels  fell  to  near 

normal  range  with  therapy  then  rose  again  when  the  patients  relapsed. 

NSE  has  been  identified  in  all  cell  lines  of  SCCL  tested;  it  has  not 

been  found  in  substantial  amounts  outside  central  and  peripheral 

nervous  system  tissue,  findings  consonant  with  the  fact  that  APUD 

cells  and  neurons  tend  to  express  much  of  the  same  genetic 

25 

information — they  are  ’’neuro-endocrine  programmed.'*  NSE  thus  has 
the  potential  to  be  a  useful  marker  for  SCCL;  iramunohistochemical 
staining  of  lung  tissue  for  NSE  might  someday  help  in  making 
pathological  diagnoses. 

A  recent  cytomorphological  finding  deserves  mention.  SCCL  cells 

from  biopsies  and  derived  cell  lines  were  shown  to  contain 

31 

neurofilament-type  intermediate  filaments.  Since  the  expression  of 
these  filaments  is  tissue  type  specific  and  thought  to  be  unchanged 
after  malignant  transformation,  another  line  of  approach  to  the  APUD 
origin  and  diagnosis  of  SCCL  may  have  been  uncovered. 

Despite  the  uncertainties  described,  a  picture  begins  to  emerge 
of  the  cells  that  make  up  SCCL  tumors.  At  a  cytogenetic  level,  they 
are  characterized  by  variable  numbers  of  chromosomes  (ploidy)  and  a 
specific  acquired  deletion  of  the  short  arm  of  chromosome  #3 — 3p 
(14-23).  The  cells  contain  neurosecretory  granules  and  appear  to  be 
of  APUD  origin.  Although  SCCL  cells  express  a  variety  of  biochemical 


13 


products,  including  certain  hormones  which  account  for  clinical 

syndromes  associated  with  the  disease,  truly  useful  biochemical 

markers  for  SCCL,  which  combine  sensitivity  and  specificity,  may 

include  neuron-specific  enolase  (NSE);  bombesin  (BN);  and  the  BB 

isoenzyme  of  creatine  kinase  (CK-BB).  Finally,  the  idea  that  SCCL  is  a 

collection  of  multiple  cell  lines,  both  in  individual  tumors  and 

between  patients,  is  supported  by  the  variety  of  tumor  products  and 

their  levels  expressed  by  cells  from  single  tumor  sites;  by  cells 

taken  from  primary  vs.  metastatic  sites;  and  by  cells  from  tumors 

found  in  different  patients.  The  notion  of  multiple  cell  lines  is 

further  supported  by  the  variable  number  of  chromosomes  (ploidy)  found 

18 

by  Vindelov  et  al .  in  some  tumor  samples  from  a  single  metastatic 

24 

site  and  by  Hattori  et  al.'s  finding  of  decreased  or  absent 

neurosecretory  granules  in  the  cells  of  tumors  unresponsive  to 

chemotherapy.  The  characteristics  of  SCCL  have  been  further 

elucidated  by  recent  studies  which  build  on  the  picture  above. 

Tumor  cell  chromosome  number  was  recently  examined  in  relation  to 

32 

treatment  response  by  a  group  at  M.D.  Anderson  Hospital.  They  found 
a  DNA  index  of  0.70  to  2.09  ( 1 . 0=diploid)  among  126  pre-treatment 
specimens.  Six  percent  of  the  cases  had  bi-clonal  stem  lines. 
Hypodiploid  tumors  had  decreased  percent  S-phase  cells  (reflecting 
lesser  proliferative  activity).  These  tumors  showed  slower  drug 
response,  but  the  response  was  more  prolonged  with  resultant  better 
survival  when  compared  to  hyperdiploid  tetraploid  tumors  characterized 
by  high  %  S-phase  cells.  Percent  survival  at  more  than  60  weeks  could 


14 


be  stratified  by  DNA  index  in  a  significant  way,  raising  the 

possibility  that  chromosome  number  and  proliferative  activity  analysis 

may  one  day  be  used  as  prognostic  criteria.  The  presence  of  more  than 

one  cell  line  in  some  tumors  was  again  confirmed. 

The  use  of  monoclonal  antibodies  (MoAbs)  may  prove  to  be  a  useful 

method  whereby  antigenic  expression — and  thus,  indirectly,  the  tumor 

genome — can  be  studied.  Implications  for  future  treatment  design  are 

numerous  and  include  the  attachment  of  anti-tumor  drugs  to  MoAbs, 

creating  a  highly  specific  tumoricidal  agent. 

Groups  at  the  NCI  have  published  numerous  abstracts  in  the  last 

two  years  reporting  on  studies  in  which  MoAbs  have  been  applied. 

Considerable  antigenic  heterogeneity  has  been  found  within  individual 

SCCL  tumors,  between  tumor  lines,  and,  to  a  more  limited  degree, 

33 

between  clonally  related  lines.  SCCL  is  thus  proving  to  be 

heterogeneous  in  a  way  that  challenges  our  understanding  of  cellular 

behavior  at  the  level  of  molecular  genetics.  Relative  homogeneity  has 

been  found  in  cell  lines  from  different  metastatic  sites  in  the  same 

patient.  This  homogeneity  was  especially  evident  when  numerous 

characteristics  of  the  cells  were  examined  simultaneously:  the 

biomarkers  dopa  decarboxylase  (DDC)  and  bombesin  (BN),  DNA  index,  and 

34 

three  forms  of  antigenic  expression  (using  MoAbs).  Still,  the 

heterogeneity  of  antigen  expression  in  tumor  tissue  and  the  poor 

specificity  of  "tumor"  antigens — i.e.,  their  diverse  distribution  in 

normal  tissue — may  cause  considerable  difficulty  in  the  clinical 

35 

application  of  MoAbs. 


15 


A  finding  which  may  prove  to  be  specific  is  that  of  HLA-A,B,C, 
and  beta-2  microglobulin  deficiencies  on  the  cell  surfaces  of  human 
SCCL  lines,  detected  by  MoAbs.  Thusfar,  non-SCCL  lines  have  been 


strongly  positive  for  these  structural  markers. 


36,37 


38  39 

Two  exceptionally  interesting  reports  from  the  NCI  ’  have 
appeared  recently  which  illustrate  our  ability  to  study  SCCL  at  a 
cellular  level  through  the  use  of  biochemical  markers,  continuous  cell 
lines,  and  measurements  of  radiosensitivity  and  tumorogenicit y .  The 
authors  report  two  major  subgroups  of  SCCL:  a  classic  form,  which 
expresses  DDC,  BN,  NSE  and  CK-BB;  and  a  variant  form,  which  has  a 
faster  doubling  time  and  shorter  latent  period  to  tumor  induction  in 
nude  mice  than  the  classic  form.  The  variant  form  is  radioresistant, 
nor  does  it  produce  DDC  or  BN  in  appreciable  quantities.  It  has 
metabolic  features  which  distinguish  it  from  the  classic  form: 
despite  the  presence  of  CK-BB,  the  product  whose  formation  that  enzyme 
catalyzes  ( phosphocreatine)  is  not  present  in  classic  cell  lines. 
Phosphocreatine  isn't  present  in  non-SCCL  lines,  but  is  present  in  the 
variant  cell  lines. 

The  possibility  that  there  are  two  major  subgroups  of  SCCL — one 

clinically  aggressive,  resistant  to  treatment,  and  lacking  the 

characteristic  APUD  enzyme  (DDC) — is  reminiscent  of  the  findings  of 

2  A 

Hattori  et  al.  Recall  that  those  authors  reported  on  the  lack  of 


neurosecretory  granules  (NSGs)  in  a  group  of  tumors  resistant  to 
chemotherapy.  Are  Hattori 's  resistant  tumors  composed  of  cells  from 
the  NCI's  variant  subgroup?  The  absence  of  DDC  in  a  tumor  one  would 


16 


expect  to  be  clinically  aggressive  (the  SCCL  variant)  is  like  the 

absence  of  NSGs  in  tumors  proven  to  be  therapy-resistant;  basic  APUD 

characteristics  are  lacking.  The  usefulness  of  APUD  characteristics 

in  understanding  SCCL  is  not  clear.  The  APUD  origin  of  some  cell 

lines  becomes  questionable.  Perhaps  tumor  cells  lacking  NSCs  and  cell 

lines  with  characteristics  of  the  variant  subgroup  are  highly 

aggressive  subpopulations  of  cells  which  have  evolved  from  the 

original,  more  indolent  APUD-derived  tumor.  The  concept  of  classic 

and  variant  cell  lines  and  the  presence  of  more  than  one  cell  line  in 

32 

a  tumor  also  brings  to  mind  the  report  from  M.D.  Anderson  which 
stratified  tumor  aggressivity  by  chromosome  ploidy  and  proliferative 
activity. 

Cytogenetic  study  of  the  classic  and  variant  cell  lines — 

evaluating  chromosome  number  and  seeking  the  deletion  in  chromosome  #3 

19 

associated  with  SCCL  — as  well  as  cytomorphological  study,  with  a 
special  interest  in  neurosecretory  granules,  may  prove  fruitful. 
Clearly,  researchers  are  only  beginning  to  explore  the  cellular  basis 
of  SCCL's  clinical  behaviors. 


Clinical  Diagnosis 

Cohen  and  Matthews, Matthews  and  Hirsch^^  and  Matthews^^  have 
reviewed  the  radiographic,  clinical,  and  pathological  presentations  of 
SCCL. 

On  X-ray,  SCCL  usually  appears  as  a  central  mass.  Because  the 
tumor  metastasizes  early,  hilar  node  involvement  is  common,  with  or 


17 


without  mediastinal  widening  on  presentation  (64%).  Post¬ 
obstructive  pneumonitis,  atelectasis,  and  pleural  effusion  (due  to 
lymphatic  blockage)  may  be  present.  Less  commonly,  a  peripheral  tumor 
mass  is  seen  on  presentation  (19%).  Very  rarely,  the  patient  presents 
with  a  central  tumor  mass  and  no  obvious  nodes  (3%).  The  lesion  must 
be  distinguished  from  epidermoid  (squamous  cell)  carcinoma,  which  also 
presents  as  a  central  lesion.  But  epidermoid  lesions  are  rarely 
associated  with  mediastinal  adenopathy  or  widening  as  evident  as  that 
in  SCCL.  Moreover,  SCCL  tumors  demonstrate  central  cavitation  less 
frequently  than  squamous  cell  lesions. 

While  the  pathogenesis  of  SCCL  and  epidermoid  cancer  is  probably 
similar,  there  are  numerous  differences  in  gross  pathology. 

Epidermoid  tumors  are  often  bulky,  polypoid,  obstructive  intraluminal 
growths,  with  a  friable  consistency,  with  or  without  central 
cavitation  and  liquefaction  necrosis.  SCCL,  in  contrast,  tends  to 
form  submucosal  plaques  that  spread  to  involve  central 
structures:  the  trachea,  mainstem  bronchi,  and  the  bronchial,  hilar, 

and  mediastinal  lymph  nodes.  If  the  superior  vena  cava  is  invaded,  it 
can  be  thrombosed,  causing  SVC  syndrome.  (SVC  syndrome  may  also  arise 
secondary  to  compression.)  SCCL  tumors  have  a  glossy  grey-white  cut 
surface  that  is  frequently  hemorrhagic  and  necrotic — but  central 
cavitation  is  rare. 

Pathological  classification  of  SCCL  is  based  on  light 
microscopy.  In  1977,  the  World  Health  Organization  revised  their 
classification  system  based  on  a  decade  of  study.  The  subtypes  now 


18 


used  include  lymphocyte-like  or  oat  cell  (#21);  intermediate  forms 
(fusiform,  polygonal,  "other” — #22);  and  combined  (oat  cell  with  a 
definite  component  of  squamous  cell  or  adenocarcinoma). 

The  nuclear  detail  of  small  cell — its  fine  or  coarse  stippled 
pattern  of  nuclear  chromatin  and  small,  indistinct  nucleoli — is  highly 
characteristic.  Cytoplasm  is  usually  scanty  or  may  appear  to  be 
absent.  Cells  are  arranged  in  a  loosely  cohesive  but  clustered 
pattern,  forming  cords,  sheets,  or  pseudorosettes  (cuffing  around 
blood  vessels). 

There  are  numerous  problems  that  arise  in  diagnosing  SCCL,  as 

40 

reviewed  by  Matthews  and  Hirsch.  These  include  inadequacy  of  biopsy 

material  (biopsy  samples  that  are  too  small,  bronchial  washings  or 

sputa  of  equivocal  cytology);  crushing  of  tissues,  resulting  in 

overinterpretation  of  malignancy;  and  improper  tissue  processing,  with 

resultant  artifacts.  The  authors  contributed  to  two  interobserver 

studies  designed  to  identify  problems  and  assess  reliability  in  SCCL 
42  43 

diagnosis.  ’  They  found  unanimity  or  "near  unanimity"  (7  of  8 
pathologists)  in  the  diagnosis  of  SCCL  in  over  90%  of  the  tumors 
studied.  The  consistency  of  subtyping  of  SCCL  tumors  according  to  the 
1977  WHO  criteria  was  assessed  in  one  of  the  studies.  Unanimity  among 
3  pathologists  was  achieved  in  only  54%  of  the  cases.  This  is  a 
remarkable  figure,  for  it  raises  doubts  about  all  studies  designed  to 
characterize  SCCL  subtypes  (e.g.,  the  response  of  different  subtypes 
to  therapy — a  topic  to  be  discussed  later). 


19 


The  great  majority  of  patients  with  SCCL  are  symptomatic  at 

presentation.  Cough  is  a  common  symptom,  referable  to  the  primary 

tumor.  Chest  pain,  dyspnea,  symptoms  of  pneumonitis  (due  to 

obstruction  or  compression),  wheeze  and  hemoptysis  may  occur.  (See 
12  44 

Table  3.)  ’  Mediastinal  extension  of  the  tumor  results  in 

hoarseness  or  SVC  syndrome,  the  former  secondary  to  involvement  of  the 

recurrent  laryngeal  nerve  (usually  on  the  left,  where  its  course  is 

longer).  SVC  syndrome  tends  to  be  associated  with  right  lung  tumors, 

since  the  superior  vena  cava  passes  through  the  chest  on  the  right. 

Early,  widespread  metastases  are  the  hallmark  of  SCCL  and 

45 

contribute  to  its  symptomatology.  Livingston  et  al.,  in  their 

series  of  375  patients,  found  liver  to  be  the  metastatic  site  most 

often  involved  in  extensive-disease  patients,  followed  closely  by 

bone;  then,  bone  marrow,  brain,  skin/soft  tissue/nodes,  and  pleural 

45 

effusion.  (See  Table  4.)  Thirty-seven  percent  of  these  patients 

had  involvement  of  more  than  one  metastatic  site,  in  contrast  to  49% 

46 

in  a  study  of  106  patients  at  the  NCI.  The  significant  metastatic 

sites,  in  terms  of  symptomatology,  are  bone  and  brain,  causing  pain 

and  neurological  complaints,  respectively.  Although  cardiac 

involvement  is  rarely  mentioned  in  clinical  series,  20-25%  of  SCCL 

patients  have  been  reported  to  have  cardiac  metastases  at 
12, 14 

autopsy.  ’  (See  Table  4.)  Such  involvement  can  result  in  signs  and 
symptoms  of  heart  failure,  EKG  changes,  even  tamponade.  Adriamycin, 
commonly  used  in  treatment  protocols  for  SCCL,  has  cardiotoxic  side 
effects;  thus,  ejection  fractions  are  routinely  computed  at  the  start 


I 


20 


of  therapy.  Still,  one  wonders  what  implications  subclinical  cardiac 

involvement  might  have  with  respect  to  treatment  complications. 

The  existence  of  clinical  syndromes  related  to  ectopic  hormone 

production  by  SCCL  tumors  has  already  been  mentioned.  All  are  rare. 

SIADH,  reported  in  5-10%  of  most  series,  seems  to  occur  most 

frequently  in  association  with  SCCL  when  the  cause  of  SIADH  is 

22 

neoplastic.  CNS  manifestations  predominate  in  symptomatic  patients 

and  may  include  seizures,  disorders  of  consciousness,  and 

extrapyramidal  signs.  The  patient  is  unable  to  excrete  a  maximally 

dilute  urine  when  presented  with  a  water  load  and  such  testing  can 

uncover  many  subclinical  cases.  Fluid  restriction  helps  correct  the 

hyponatremia,  but  chemotherapy  is  the  definitive  approach. 

Ectopic  ACTH  production  is  clinically  significant  in  3-7%  of 

22 

patients  with  SCCL.  Nmnerous  tumors  thought  to  be  of  APUD  origin 

make  the  hormone.  Symptomatic  patients  rarely  present  with  the 

classic  features  of  Cushing's  syndrome — instead,  weight  loss,  severe 

weakness,  glucose  intolerance,  edema  and/or  hypertension  are  more 

likely  presentations  of  their  hypercortisolism.  The  metabolic 

complications  of  symptomatic  ACTH  overproduction  can  be  severe  and 

22 

management  is  difficult,  although  Greco  et  al.  report  early  evidence 
that  combination  chemotherapy  can  be  effective. 

Paraneoplastic  syndromes  other  than  ectopic  hormone  production 
have  been  found  to  be  associated  with  SCCL.  Possible  etiologies  of 
these  syndromes  include  viral  agents,  autoimmune  phenomena,  and 
humoral  substances  elaborated  by  the  tumor. 


21 


Eaton-Lambert  syndrome  is  associated  with  SCCL  more  often  than 

22 

with  other  diseases.  It  is  an  unusual  clinical  entity, 
characterized  by  the  dichotomous  findings  of  proximal  muscle  weakness 
with  difficulty  walking  coupled  with  facilitation  of  muscular 
potentials  on  repeated  stimulation.  Thus,  the  patient's  grip  may 
become  stronger  and  stronger  during  testing.  The  syndrome  tends  to 
occur  among  male  patients  over  40  years  of  age  and  seems  to  respond  to 
cytotoxic  therapy.  Should  chemotherapy  fail,  guanidine  may  be 

22 

effective  by  causing  increased  release  of  anticholinesterases.  The 

electromyogram  is  diagnostic. 

A  final  syndrome  requiring  mention  is  paraneoplastic 

22 

encephalopathy.  This  is  thought  to  be  the  cause  of  death  in  two 
patients  in  the  present  study.  Clinically,  the  syndrome  may  involve 
the  cerebrum,  brainstem,  optic  nerves,  and  cerebellum.  Pathologic 
lesions  are  generally  found  in  all  these  regions,  although  involvement 
of  one  area  may  dominate  the  clinical  picture.  Dementia  is  the  most 
common  manifestation  of  cerebral  involvement.  Radiologic  studies  are 
normal;  the  CSF  may  show  an  elevated  protein  level;  the  E<EG  is  often 
slow  and  diffuse. 


Staging  and  Prognosis 

There  is  a  TI#1  ( tumor-nodes-metastases)  staging  system  for  SCCL 
but  its  use,  until  recently,  had  fallen  out  of  favor.  The  TNM  system 
is  surgically-oriented  and  poor  therapeutic  results  have  been  achieved 
using  surgery  alone.  TNM  was  viewed  as  prognostically  useless.  More 


22 


recently,  however,  interest  in  surgery  as  an  adjuvant  therapy  (part  of 
a  multimodal  therapeutic  approach)  has  been  revived  and  the  TNM  system 
may  yet  take  its  place  as  a  standard  method  of  staging.  (See  the 
section  on  treatment.)  Still,  the  system  of  the  Veterans 
Administration  Lung  Cancer  Study  Group  remains  the  one  employed  in 
almost  all  current  studies.  It  divides  patients  into  limited-stage 
(LD)  and  extensive-stage  (ED)  disease  groups.  LD  is  defined  as  tumor 
confined  to  one  hemithorax  with  or  without  mediastinal 
lymphadenopathy ,  with  or  without  ipsilateral  supraclavicular  node 
involvement.  The  tumor  must  fit  within  a  single  radiation  therapy 
portal.  Tumor  beyond  these  confines  is  defined  as  extensive 
disease. 

In  general,  two-thirds  of  all  patients  present  with  ED;  one-third 

5  48  49  48 

with  LD.  ’  ’  Ihde  and  Hansen  have  pointed  out  that,  with  very 

thorough  diagnostic  work-ups,  more  patients  with  metastatic  disease 
not  easily  detectable  are  placed  in  the  ED  group.  When  this  is  done, 
survival  data  for  the  individual  ED  and  LD  groups  may  appear  improved, 
since  a  group  of  relatively  "healthy”  ED  patients  is  created  by 
removing  a  group  of  relatively  "sick"  LD  patients.  Overall  survival 
(ED  +  LD)  does  not  change.  The  truth  of  this  observation  must  be 
supplemented  by  the  observation  that,  despite  the  sophistication  of 
the  technology  available  to  the  physician  determined  to  uncover  the 
most  retiring  metastasis,  test  results  are  often  equivocal.  It  is  not 
clear  whether  or  not  to  include  certain  patients  in  the  ED  group  and 
they  may  be  given  the  "benefit  of  the  doubt" — identified  as  patients 


A 


23 


with  limited  disease  so  that  they  might  enter  the  LD  treatment  group. 
When  this  happens,  the  survival  of  the  ED  group  presumably  goes  up  and 
that  of  the  LD  group  goes  down.  Again,  overall  survival  remains 
unchanged . 

Involvement  of  certain  metastatic  sites  has  been  found  to  have 
prognostic  significance.  More  fundamentally,  extent  of  disease  has 
strong  prognostic  implications.  Staging  procedures  in  SCCL  must  be 
designed  with  these  facts  in  mind.  Diagnostic  modalities  must  be 
selected  for  their  combination  of  sensitivity  and  specificity. 

Chest  X-ray  remains  the  basic  method  by  which  intrathoracic 
disease  is  evaluated.  However,  fiberoptic  bronchoscopy  is  a  very 
useful  tool — it  allows  diagnosis  by  biopsy  or  bronchial  washings  and 
can  detect  small  lesions.  The  bronchoscope  is  used  routinely  at  some 
centers  to  document  complete  response  to  therapy. 

CT  scans  of  the  chest  are  less  widely  used,  although  they  provide 

a  better  sense  of  tumor  volume.  Some  note  that  the  CT  scan's 

sensitivity  is  not  matched  by  its  specificity  in  detecting  malignant 

pulmonary  nodules  when  compared  to  conventional  linear 

tomography . Others  point  to  the  relative  inaccuracy  of  CT  scans  in 

diagnosing  disease  in  the  middle  mediastinum  when  compared  to  staging 

mediastinoscopy  or  thoracotomy.^^  Still,  CT  provides  useful 

51  52 

information  not  obtained  with  conventional  radiography.  ’ 

Both  mediastinosocopy  and  CT  of  the  chest  may  take  on  a  more 


important  role  in  initial  staging  in  the  future.  The  reason  for  this 
is  the  resurgence  of  interest  in  TNM  staging  to  evaluate  adjuvant 


24 


surgery  for  early  control  of  intrathoracic  tumor  mass.  Surgery  aside, 

data  is  accumulating  that  would  indicate  a  survival  advantage  for 

patients  with  smaller  presenting  tumors  in  the  chest.  A  group  in 
53 

England  found  a  higher  incidence  of  complete  response  and  a  survival 

advantage  for  patients  with  intrathoracic  tumors  whose  total 

2  54 

cross-sectional  was  area  less  than  30  cm  on  CT.  A  Toronto  group 

reported  a  significant  survival  advantage  for  limited  disease  patients 

without  superior  mediastinal  node  involvement  as  diagnosed  by 

mediastinoscopy  or  roentgenographic  appearance — i.e.,  patients  with 

so-called  "very  limited"  disease,  which  is  potentially  resectable. 

These  two  studies  illustrate  the  potential  use  of  CT  and/or 

mediastinoscopy  in  evaluation  of  chest  tumor  extent.  Such  information 

could  be  useful  for  treatment  and/or  prognosis. 

Bone  and  bone  marrow  are  common  sites  of  metastatic  spread.  It 

seems  important  to  perform  both  bone  marrow  biopsy  and  aspiration^^  as 
56 

well  as  bone  scan  if  extent  of  disease  is  to  be  assessed,  for  the 


procedures  complement  each  other  to  some  extent.  The  prognostic 

57 

significance  of  these  sites  of  involvement  is  unclear,  however.  Two 

recent  studies  found  bone  marrow  involvement  to  be  a  negative 

58  59 

prognostic  factor;  ’  a  third  report  found  no  prognostic 

significance.^^  A  study  from  the  Finsen  Institute^^  identified  a 

group  with  an  especially  poor  prognosis:  patients  with  bone  marrow 

metastases  and  thrombocytopenia.  In  any  case,  bone  scan  and  bone 

marrow  biopsy  and  aspiration  remain  standard  procedures  in  staging 

48 

SCCL.  Ihde  and  Hansen  have  reported  worsening  of  the  bone  scan 


25 


during  overall  disease  remission  in  a  minority  of  patients. 

Liver  metastases  appear  to  be  a  negative  prognostic  factor. 

46  61  59 

Groups  at  the  NCI  ’  and  Toronto  have  found  liver  involvement  to 

bode  ill  and  the  level  of  interest  in  accurate  diagnosis  of  liver 

metastases  supports  the  wide  acceptance  of  this  notion. 

A  variety  of  techniques  may  be  used  to  evaluate  the  liver, 

including  liver  function  tests,  radionuclide  scanning,  CT, 

peritoneoscopy  with  biopsy,  percutaneous  biopsy,  and  ultrasonography 

with  fine  needle  aspiration.  Liver-spleen  radionuclide  scan  remains 

the  mainstay  of  current  staging.  Although  this  modality  has  been 

maligned,  it  remains  a  reliable,  available,  relatively  inexpensive, 

62 

non-invasive  diagnostic  tool  in  SCCL.  At  the  NCI,  peritoneoscopy 

with  liver  biopsy  was  found  to  be  the  most  sensitive  diagnostic 

method,  but  an  algorithm  combining  radionuclide  scan  with  liver 

function  tests  was  highly  accurate  while  remaining  non-invasive.^^  A 

63 

recent  study  from  the  Finsen  Institute  reported  ultrasonography  with 
fine  needle  aspiration  to  be  more  accurate  than  peritoneoscopy.  The 
ultrasound  technique  was  also  ’’less  invasive" — but  the  number  of 
patients  studied  was  small. 

Whether  or  not  prophylactic  irradiation  should  be  employed  to 
avoid  CNS  metastases  in  patients  with  SCCL  is  a  controversial  topic. 
Less  controversial  is  the  prognostic  significance  of  CNS  involvement 
and  how  to  diagnose  it. 

Brain  metastases  are  present  in  approximately  10%  of  patients 

48 


with  SCCL  at  diagnosis  and  in  30-65%  of  patients  at  autopsy. 


As 


26 


therapy  improves  and  patients  with  SCCL  live  longer,  they  accumulate 

greater  risk  for  developing  this  complication. 

Both  the  NCl'^^  and  Toronto^^  groups  found  brain  metastases  to  be 

a  negative  prognostic  factor,  but  this  result  has  not  been 

invariable. Diagnosis  can  be  made  by  radionuclide  scan  or  CT  of  the 

head.  CT  scans  are  thought  to  be  superior, uncovering  lesions 

64 

before  they  become  symptomatic  and  allowing  accurate  staging. 

A  current  set  of  recommendations  for  restaging  of  patients 

thought  to  have  achieved  clinical  response  is  presented  in  Table 

5.^^  The  argument  that  restaging  should  be  effected  with  as  much  care 

as  initial  staging  is  a  good  one  since  any  metastasis  large  enough  to 

be  clinically  detectable  is  of  great  significance.  Perhaps  someday 

biomarkers  already  described  (e.g.,  bombesin,  neuron-specific  enolase) 

will  become  standard  indicators  of  subtler  disease. 

What  factors  can  be  said  to  have  prognostic  significance  in  SCCL? 

Performance  status — a  numerical  estimate  of  a  patient's  ability  to  go 

about  his  daily  routine  with  or  without  symptoms — and  stage  of  disease 

(limited  vs.  extensive),  predominate  in  most  studies  of  significant 

48  59 

prognostic  factors.  ’  In  fact,  the  International  Association  for 

the  Study  of  Lung  Cancer  feels  it  is  no  longer  necessary  to 

demonstrate  the  superior  survival  of  patients  with  LD  in  current 

chemotherapy  treatment  reports. Still,  significantly  improved 

AG 

survival  for  LD  patients  is  not  a  universal  finding.  Data  already 
mentioned  indicate  that  small  intrathoracic  tumor  area  and  a  finding 
of  "very  limited"  (i.e.,  potentially  resectable)  disease  may  confer  a 


27 


53  54 

prognostic  advantage.  ’  Patients  who  have  failed  a  previous 

chemotherapeutic  protocol  invariably  have  a  bad  prognosis  on 

48 

second-line  therapy. 

The  following  are  of  less  certain  significance.  Weight  loss  on 

presentation  (0-10%)  has  been  found  to  be  associated  with 

66 

significantly  decreased  median  survival,  as  has  multiple  metastatic 

46 

sites  (one  vs.  two  vs.  three  or  more).  Age  of  55  years  of  more  has 
been  associated  with  decreased  response  rates  and  shorter  survival  in 
patients  not  achieving  complete  response. 

Numerous  laboratory  parameters  may  be  useful  as  prognostic 
indicators;  carcinoembryonic  antigen  (CEA)  has  been  reported  to  be  an 
independent  prognostic  factor, as  has  The  feasibility  of 

using  an  objective  prognostic  index  based  on  laboratory  parameters  at 
diagnosis  to  replace  subjective  performance  status  assessment  is  under 
study.  At  the  NCI,^^  albumin  and  hemoglobin  were  found  to  be  the  most 
influential  prognostic  factors  in  survival. 

The  fact  that,  of  the  various  metastatic  sites,  CNS  and  liver  are 
the  most  likely  to  have  prognostic  significance  has  already  been 
discussed. 

Finally,  the  prognostic  significance  of  the  various  subtypes  of 
SCCL  remains  unclear.  The  VA  Lung  Group^^  reported  better  survival 
for  patients  with  lymphocyte-like  (classic,  oat-cell) 

vs.  intermediate-type  disease.  This  held  true  for  patients  within  the 

extensive  disease  group,  but  not  the  limited  disease  group,  when 

72 

analyzed  separately.  In  contrast,  a  large  NCI  study  found  no 


■5 


J 


28 


clinically  significant  differences  among  the  subtypes.  A  recent 

community  hospital  study  indicates  that  lymphocyte-like  SCCL  may  be 

73a 

associated  with  better  2  year  survival. 

The  pathology  panel  of  the  International  Association  for  the 

Study  of  Lung  Cancer  is  in  the  process  of  proposing  a  revision  of  the 

73b 

current  WHO  histological  classification  system  for  SCCL.  The 

"combined"  subgroup  would  remain;  however,  "oat  cell"  and 
"intermediate"  subtypes  would  be  classified  together  in  the  new 
"classic  small  cell"  subgroup.  Another  new  subgroup  would  be 
created:  "small  cell-large  cell,"  in  which  there  is  an  admixture  of 

classic  small  cells  with  large  cells  having  open  nuclei  and  prominent 
eosinophilic  nucleoli. 

73b 

Dr.  Raymond  Yesner  has  reported,  in  a  personal  communication, 
that  the  new  classification  system  is  based  on  the  belief  that 
polygonal  and  fusiform  cell  types — currently  grouped  in  the 
intermediate  subtype — show  no  significant  clinical,  biological,  or 
ultrastructural  differences  from  classic  oat  cells.  The  VA  Lung  Group 
study, as  reported  earlier  in  this  paper,  found  a  survival  advantage 
for  oat  cell  over  intermediate-type  disease.  It  turns  out  that  this 
group,  unlike  investigators  who  found  no  such  survival  advantage, 
included  in  the  intermediate  subtype  only  tumors  with  a  mixture  of 
classic  small  cells  and  large  cells — not  tumors  of  polygonal  and 
fusiform  cells,  which  they  grouped  with  classic  oat  cell  tumors.  The 
proposed  classification  system  is  based  on  the  belief  that  tumors  of 
the  small  cell-large  cell  subgroup  carry  a  graver  prognosis  than  those 


29 


of  the  classic  small  cell  subgroup. 

Finally,  the  new  small  cell-large  cell  subgroup  is  thought  to  be 

identical  to  the  vitro  "variant"  cell  line  described  earlier  in 

38  39 

this  paper  based  on  reports  from  the  NCI.  ’  The  variant  cell  line 
is  relatively  resistant  to  radiation  and  chemotherapy  and  has  a  large 
cell  appearance,  but  produces  some  characteristic  SCCL  biomarkers. 
Similarly,  the  classic  small  cell  subgroup  is  felt  to  have  its  in 
vitro  equivalent  in  the  NCI’s  "classic"  subclass. 

Table  6  presents  a  summary  of  prognostic  factors. 


PART  TWO:  TREATMENT 


Overview 

A  better  theoretical  grasp  of  cancer  in  general  now  permits 
clinicians  to  treat  SCCL  with  a  modest  degree  of  success.  As  the 
characteristics  of  the  tumor  become  better  understood,  the  limitations 
of  current  therapy  become  painfully  clear.  Will  the  treatment 
breakthrough  come  from  the  slow  evolution  of  rational  therapeutic 
design  or  through  a  fortuitous  discovery?  It  is  unlikely  that 
stepwise  investigation  will  cure  SCCL  in  the  near  future;  and,  given 
the  cost-conscious  environment  of  present-day  cancer  research,  the 
prospect  of  a  serendipitous  discovery  is  better  thought  of  as  a 
fantasy  than  a  hope. 

In  order  to  treat  SCCL  and  to  evaluate  properly  its  response  to 
treatment,  the  growth  characteristics  of  the  tumor  need  to  be  known. 
Two  basic  approaches  exist:  measures  of  clinical  doubling  time  made 
by  estimating  tumor  volume  changes  on  chest  radiographs  over  time;  and 
in  vitro  studies  of  tumor  cells.  Tritiated  thymidine  uptake  by  SCCL 
cells  allows  calculation  of  the  labeling  index  (LI) — the  fraction  of 
labeled  tumor  cells  among  all  cells  counted.  LI  reflects  the  rate  of 
cell  production  by  the  tumor — it  measures  the  fraction  of  cells 
actively  synthesizing  DNA. 

SCCL  responds  well — initially — to  treatment  with  radiation 
73c 

therapy  (which  acts  on  dividing  cells)  and  to  treatment  with  agents 
such  as  methotrexate  (which  is  S-phase  specific)  and  cyclophosphamide 


31 


74 

(which  is  probably  cycle-dependent).  Intuitively,  one  would 

therefore  expect  SCCL  to  be  a  rapidly  dividing  tumor — one  with  a  high 

LI  and  short  doubling  time  as  measured  in  clinical  studies. 

Initial  results  were  consistent  with  this  view.^^  For  a  while, 

researchers  took  a  value  of  about  one  month  as  the  doubling  time  of 

small  cell  tumors.  Based  on  this  assumption,  the  period  of  risk — the 

amount  of  time  for  regrowth  from  a  single  cell  to  clinical 

recurrence — was  thought  to  be  about  two  years. 

Two  years  became  a  magical  interval,  synonymous  with  long-term 

survival^^ ’ and,  perhaps,  cure.  This  view  was  consistent  with 

clinical  impression,  doubling  time  data,  and  the  finding  that  SCCL  had 

a  higher  median  LI  than  other  solid  tumors  (except  Burkitt’s 
78 

lymphoma).  Although  it  has  since  become  clear  that  two  year 

survival  is  of  limited  value,  it  remains,  for  many,  an  important 

criterion.  A  review  article  from  1982  states  that  "many  patients  who 

survive  alive  and  disease-free  for  2  years,  remain 
79 

disease-free."  The  reference  given,  from  1979,  was  employed 

K  77 
above . 


In  1978,  a  group  at  the  NCI  reported  a  median  doubling  time  of 
77  days  (range:  25-160  days)  among  their  12  cases  of  SCCL.  Most 
tumors  were  felt  to  have  demonstrated  relatively  intermediate  or  long 
doubling  times.  Assuming  that  the  range  of  1x10^  to  1x10^  cells  is 
clinically  significant,  the  authors  used  the  median  doubling  time  of 
77  days  to  project  that  therapy  leaving  a  tumor  burden  of  1x10  cells 
would  not  present  as  a  clinical  relapse  for  at  least  two  years; 


32 


therapy  destroying  all  but  one  cell  would  produce  an  interval  of  risk 
lasting  4-5  years. 

The  above  projections  may  not  be  entirely  accurate.  First,  the 

range  of  tumor  doubling  times  must  be  taken  into  account.  Second, 

treatment  may  change  the  cell  kinetic  characteristics  of  SCCL 
78 

tumors;  there  is  good  evidence  to  support  biochemical  and 

81  82 

histological  changes  in  tumors  after  therapy.  ’  Still,  4  or  5  year 
survival  is  probably  more  accurately  synonymous  with  long-term 
survival  if  cure  is  implied.  Clinical  evidence  has  accumulated  to 
support  this  notion  in  the  form  of  late  relapses. 

Such  evidence  has  been  available  for  a  number  of  years.  The 

83 

NCI-International  Association  for  the  Study  of  Lung  Cancer  report 

which  appeared  in  1980  studied  patients  who  survived  more  than  2.5 

years.  Recurrent  disease  was  noted  in  21  of  96  patients:  8  patients 

died  30-33  months  after  diagnosis;  one  was  alive,  with  disease,  at  34 

months;  recurrence  was  detected  in  10  other  patients  after  36-51 

months;  and  two  patients  treated  by  surgery  alone  succumbed  to 

recurrent  disease  at  8  and  9  years,  respectively. 

Recently,  data  from  cooperative  and  single  institutions  have  been 

84 

gathered  on  late  relapses.  The  NCI  reported  that  8  of  28  patients 
who  had  been  disease-free  at  30  months  relapsed  with  SCCL  (median:  54 
months  from  diagnosis;  range:  31-74  months)  after  follow-up  of  5-10 
years.  The  group  at  M.D.  Anderson  Hospital  reviewed  patients 
surviving  3  years  or  more.  Eleven  of  43  such  patients  relapsed 
systemically .  Seven  of  the  11  relapses  were  at  more  than  3 


33 


85  86 

years.  Livingston,  reporting  for  the  Southwest  Oncology  Group, 

looked  at  17  patients  who  survived  5  years  or  more  in  a  single  study 

(13%  of  those  entered).  Five  late  deaths  were  due  to  recurrent  tumor 

(onset:  33-73  months  from  treatment). 

87 

As  early  as  1978,  Greco  et  al.  published  a  paper  in  which  the 

notion  that  2  year  survival  might  not  represent  cure  was  clearly 

expressed.  The  existence  of  "late  recurrences"  was  recognized. 

Before  discussing  treatment  modalities:  What  is  the  natural 

history  of  SCCL?  The  median  length  of  survival  of  untreated  patients 

with  SCCL  is  generally  quoted  as  2-3  months,  depending  on  extent  of 

disease  at  presentation.^  One  widely  quoted  study  is  that  of  the  VA 
47 

Lung  Study  Group,  in  which  38  SCCL  patients  with  limited  disease 

achieved  a  median  survival  of  11.7  weeks  and  108  patients  with 

extensive  disease  achieved  a  median  survival  of  5.0  weeks  on  placebo. 

88 

In  a  cooperative  VA  study  reported  by  Roswit  et  al., 
placebo-treated  SCCL  patients  with  limited  disease  had  a  median 
survival  of  more  than  16  weeks. 

Surgery  was  one  of  the  first  modalities  used  to  treat  SCCL.  The 

results  were  not  good.  Even  apparently  resectable  lesions  were 

frequently  found  to  have  seeded  distant  sites;  relapse  was  the  rule. 

A  study  of  pathology  material  from  the  tumors  of  19  patients  who  died 

within  30  days  of  apparently  successful  surgical  resection  found 

persistent  disease  in  13  of  19  patients,  12  of  whom  had  distant 
89 

metastases.  Radiation  therapy  (RT)  alone  proved  better  than  surgery 

90 

in  a  British  Medical  Research  Council  trial.  The  patients  had 


limited  disease  thought  to  be  resectable  and  were  fit  enough  for 
surgery  or  radical  RT.  At  10  year  follow-up,  the  surgery  group  had  a 
mean  survival  of  199  days,  the  RT  group  300  days.  Three  patients  in 
the  RT  group  who  survived  five  years  remained  alive  and  disease-free 
at  10  years.  Four  RT  patients  died  between  2  and  5  years.  The  sole  5 
year  survivor  in  the  surgery  group  in  fact  underwent  no  surgical 
treatment  due  to  breathlessness  and  received  RT  instead. 

88 

In  contrast,  in  a  cooperative  VA  study  already  mentioned,  there 

was  no  significant  increase  in  survival  for  a  group  with  limited 

disease  receiving  4-5,000  rads  of  RT  compared  to  a  placebo  group. 

Median  survival  for  the  RT  group  was  a  bit  over  16  weeks. 

74 

Selawry,  in  a  1973  report,  reviewed  the  response  of  SCCL  to 

single  agent  chemotherapy.  Small  cell  was  found  to  be  the  most 

responsive  to  single  agents  of  all  lung  cancer  subtypes. 

Therapeutic  design  moved  quickly  once  the  efficacy  of 

chemotherapy  had  been  shown.  Radiation  therapy  was  combined  with 

91 

chemotherapy,  creating  a  multi-modal  approach;  multiple 

87  92 

chemotherapeutic  agents  were  employed.  ’  It  became  clear  that 

patients  who  achieved  complete  response  lived  longer  than  those 

achieving  partial  response  or  no  response  (in  complete  responders,  the 

disease  had  been  made  clinically  undetectable);  and  partial  responders 

93 

seemed  to  live  longer  than  non-responders. 

Chemotherapy  has  become  the  backbone  of  therapeutic  approaches  to 
SCCL.  Basic  principles  of  chemotherapy  design  have  been  applied  to 
the  disease.  Attempts  have  been  made  to:  combine  drugs  which  have 


35 


therapeutic  efficacy  as  single  agents;  choose  combinations  of  drugs 
with  different  modes  of  action;  treat  with  apparently  non-cross- 
resistant  sequential  combinations  of  drugs;  find  combinations  of  drugs 
with  synergistic  anti-tumor  effects;  and  use  drug  dosages  high  enough 
to  maximize  dose-response  advantages  while  minimizing  the  inherent 
trade-off  of  dose-related  toxicity. 

Disease  extent  is  a  major  prognostic  factor  in  SCCL  and  treatment 

results  reflect  this  fact.  It  is  wise  to  discuss  therapy  of  limited 

disease  and  extensive  disease  separately.  In  general,  limited  disease 

treatment  has  changed  little  in  the  past  five  years  and  is  dominated 

by  controversies  over  the  use  of  radiation  therapy  to  the  chest  and 

prophylactic  cranial  irradiation  as  adjuvants  to  combination 

chemotherapy.  An  exception  to  this  statement  is  the  renewed  interest 

in  surgery  as  an  adjuvant  therapy  in  resectable  lesions.  The  more 

creative  approaches  to  therapy — new  drugs,  larger  doses, 

non-cross-resistant  sequential  combinations — have  been  confined 

largely  to  treatment  of  extensive  disease  or  patients  who  have 

relapsed.  The  reason  for  this  is  that  current  conservative 

therapeutic  designs  produce  a  predictable,  although  small,  number  of 

long-term  survivors  in  the  limited  disease  group;  the  extensive 

93 

disease  group  has  fewer  responders  and  shorter  survival.  Clinicians 

are  reluctant  to  give  up  ’’acceptable"  survival  and  known  toxicity 

risks  for  experimental  therapies. 

65 

Aisner  et  al.,  reporting  for  the  International  Association  for 
the  Study  of  Lung  Cancer  have  summarized  current  expectations  in 


36 


trials  employing  aggressive  therapy  against  SCCL.  Combination 
chemotherapy  should  produce  complete  response  in  more  than  50%  of 
patients  with  limited  disease  (LD)  and  more  than  20%  of  patients  with 
extensive  disease  (ED).  With  adequate  staging,  median  survival  of  at 
least  14  months  in  LD  and  7  months  in  ED  may  be  expected.  Finally, 
15-20%  of  LD  patients  should  achieve  disease-free  survival  of  3  years 
or  more  although  such  survivors  remain  rare  among  ED  patients. 

Table  7  presents  a  summary  of  selected  treatment  protocols  for 
SCCL.  It  is  intended  to  show  the  evolution  of  therapy  and  variability 
of  treatment  results.  It  does  not  present  highly  experimental 
approaches  of  the  kind  usually  reserved  for  extensive  disease  patients 
or  patients  who  have  relapsed  from  first-line  therapy.  Unless  drawn 
from  the  same  paper,  the  studies  are  not  comparable. 

Table  8  presents  information  on  long-term  survivors  from  studies 
using  various  treatment  modalities. 

Treatment  of  Limited-Stage  Disease  and 
Treatment-Related  Toxicities 

Two  controversial  aspects  of  therapy  design  are  especially 
relevant  to  limited  disease,  since  their  goal  is  prophylaxis  or  rapid, 
effective  local  control:  the  use  of  prophylactic  cranial  irradiation 
(PCI);  and  intrathoracic  irradiation  for  local  tumor  control,  both  as 
adjuvants  to  combination  chemotherapy. 

Neither  non-randomized  nor  randomized  trials  of  PCI  have 

99, 103 


demonstrated  any  clear  advantage  in  survival. 


.1 


37 


99 

Baglan  and  Marks  thus  argued  that  the  nominal  purpose  of  PCI 
was  to  prevent  neurological  signs  and  symptoms,  since  their  review  of 
the  literature  uncovered  an  incidence  of  brain  metastases  averaging 
23%  for  patients  not  receiving  PCI  versus  5%  for  the  PCI  group.  The 
authors  were  able  to  treat  64%  of  39  patients  with  brain  metastases 
(all  but  4  of  whom  were  symptomatic)  successfully  enough  to  eradicate 
symptoms  for  the  rest  of  the  patients'  lives.  The  authors  predicted 
that,  based  on  their  results  treating  symptomatic  patients  and  on 
previous  treatment  results  with  PCI,  of  100  prophylactically 
irradiated  and  100  symptomatically  irradiated  patients,  77  extra 
patients  would  have  to  receive  PCI  so  that  3  patients  might  be  spared 
CNS  symptoms.  They  considered  the  potential  benefit  of  PCI  to  be 
insignificant . 

Baglan  and  Marks's  argument  hinges  on  effective  treatment  of  CNS 

metastases.  Agreement  on  this  point  is  not  uniform; still,  a 

recent  NCI  study  indicated  that  brain  metastases  can  be  treated 

effectively  enough  so  that  such  patients  die  of  other  causes  in  most 
102 

cases. 

A  large  retrospective  NCI  study^^*^  examined  PCI  with  a  special 
interest  in:  PCI  timing;  PCI's  effect  on  long-term  survival;  and 
selection  of  any  subgroups  of  patients  for  whom  PCI  would  be  most 
helpful.  The  results  were  of  great  interest:  there  was  significant 
improvement  in  overall  survival  for  the  group  receiving  PCI.  However, 
the  group  which  received  no  PCI  also  had  the  least  intensive 
chemotherapy.  With  that  caveat  in  mind,  the  authors  felt  that  PCI  had 


38 


its  greatest  positive  effect  in  the  complete  responders  (with  limited 

or  extensive  disease).  Among  patients  achieving  complete  response  who 

had  received  no  PCI,  17%  relapsed  in  a  CNS  site  alone.  Isolated  CNS 

relapse  was  seen  in  no  complete  responders  who  had  received  PCI.  Two 

and  three  year  survival  was  improved  in  the  PCI  groups,  but  not 

significantly  so.  With  respect  to  the  timing  of  PCI:  there  were  no 

CNS  relapses  in  the  first  four  months  in  any  group  and  no  striking 

treatment  result  differences  between  a  group  receiving  PCI  on  day  1  of 

the  protocol  and  a  group  receiving  PCI  at  week  12  or  24,  contingent  on 

a  complete  or  partial  therapy  response. 

The  NCI  group  thus  suggested  that  PCI  may  be  most  effectively 

employed  at  2-4  months,  after  documented  complete  response  has  been 

achieved.  Patients  achieving  less  than  complete  response  could  be 

treated  symptomatically  since  the  study  found  no  apparent  advantage 

103 

using  PCI  in  that  group.  A  recent  study  from  Toronto  found  no 
increased  survival  but  significantly  decreased  brain  relapse  at  2 
years  for  complete  responders  receiving  PCI  (21%  vs.  52%). 

Data  is  accumulating  to  support  selective  use  of  PCI — the  data  is 
on  toxicities  associated  with  PCI.  Numerous  groups  have  reported 
neurological  toxicities  among  long-term  survivors  which  may  be  due  to 

PCI  or  the  combination  of  PCI  and  chemotherapy  (nitrosureas,  in 

84,104,105  ,  ^  .  .  106  . 

particular).  A  group  at  Indiana  University  found 

neurologic  problems  in  9  of  11  long-term  disease  free  patients  (>3 

years)  who  had  received  PCI  +  nitrosureas,  and  in  6  of  8  patients  who 

had  received  PCI  and  chemotherapy  without  nitrosureas.  Onset  of 


39 


neurological  symptoms  was  usually  1-3  years  after  completion  of 
therapy.  Problems  encountered  included  memory  loss,  dementia, 
confusion,  ataxia,  psychomotor  retardation,  dysphonia  and  optic 
atrophy.  Two  patients  required  institutionalization;  6  others  have 
had  great  impairment  of  their  daily  lives.  Only  4  of  18  patients  have 
had  no  neurologic  impairment  after  therapy.  Recently,  a  prospective 
evaluation  revealed  an  '’extraordinary  high  frequency  of  CCT 
(computerized  cranial  tomography)  abnormalities  in  patients  with  SCCL 
after  treatment  with  chemotherapy  and  cranial  irradiation. 

The  role  of  PCI  in  the  treatment  of  SCCL  remains  unclear.  It 
appears  that  PCI  may  offer  a  relapse  protection  advantage  in  patients 
achieving  a  complete  response  that  is  worth  the  risk  of  possible 
long-term  neurological  side  effects.  Much  may  depend  on  the  side 
effects  clinicians  are  willing  to  tolerate  to  protect  the  subgroup  of 
complete  responders  who,  without  PCI,  would  experience  isolated  CNS 
relapse.  Neurological  side  effects  need  to  be  further  studied  so  that 
especially  toxic  PCI-chemotherapy  combinations  can  be  avoided.  More 
data  are  needed  on  survival,  relapse,  and  toxicity  through  randomized 
trials  of  PCI  in  complete  responders. 

The  controversy  surrounding  the  use  of  radiation  therapy  to  the 
chest  to  complement  multi-agent  chemotherapy  is  a  complex  one. 

Multi-modal  therapy,  referring  to  combined  radiation  and 
chemotherapy,  is  of  two  main  types:  sequential  and  concurrent.  In 
sequential  therapy,  there  is  a  temporal  pause  between  the  two 
modalities;  in  concurrent  therapy,  they  are  given  simultaneously. 


I 


'■ii 


40 


1  Oft 

Catane  et  al. ,  found  a  trend  favoring  concurrent  therapy  over 
sequential  therapy  for  increased  two  year  survival.  The  difference 
was  not  statistically  significant,  however.  The  concurrent  therapy 
group  achieved  better  complete  therapy  response  with  local  tumor 
control  and,  of  patients  achieving  complete  response,  fewer  patients 
receiving  concurrent  therapy  relapsed  in  the  radiation  therapy 
portal . 

The  toxicity  enhancement  effects  of  concurrent  therapy  are 

critical  in  evaluation  of  protocol  design.  In  Catane' s  study,  7  of  14 

patients  receiving  maximal  concurrent  radiation  and  chemotherapy  (9 

weeks)  died  of  treatment  toxicity.  Yet,  4  of  the  7  treatment 

survivors  achieved  2  year  survival — the  highest  proportion  of  any 

group  in  the  study.  The  authors  concluded  that  3  weeks  of  concurrent 

radiation  therapy  (RT)  and  chemotherapy  (CT)  produced  the  optimal 

combination  of  high  2  year  survival  and  acceptable  toxicity. 

109 

Cox  et  al.,  found  that  tumor  control  probability,  assessed  by 
serial  chest  radiographs,  increased  with  increasing  biological  dose  in 
patients  treated  with  RT  alone.  But  in  RT  +  CT  patients,  local 
control  was  achieved  at  lower  RT  doses  than  would  have  been  expected. 
RT  was  generally  begun  during  the  last  week  or  immediately  after 
completion  of  chemotherapy. 

The  point  is  that  RT  and  CT  appear  to  act  synergistically :  they 
enhance  each  other's  treatment  effects  but  they  also  enhance 
toxicities.  Acute  toxicity  enhancement  effects  include  myocardial, 
pulmonary,  skin  and  esophageal  damage  with  Adriamycin; 


chronic 


■fiki) 


i 


i 


41 


toxicities  will  be  discussed  shortly.  However,  delay  of  one  week 
between  modalities  is  thought  to  be  protective. 

We  enter  the  RT  +  CT  vs.  CT  alone  controversy  with  this 
perspective:  the  timing  of  combined  modality  treatment  is  important 

for  toxicity  and  anti-tumor  effects;  RT  seems  to  act  synergistically 
with  CT  on  tumor  cells.  To  date,  the  critical  parameters  of  RT-CT 
combination  therapy — timing  and  dosage — have  not  been  adequately 

j  111 

studied. 

The  main  argument  for  combined  modality  treatment  is  local  tumor 

112 

control.  Byhardt  and  Cox  argue  that  failure  of  chemotherapy  alone 

to  prevent  relapses  in  the  chest  is  the  reason  to  add  adjuvant  RT. 

Combined  modality  therapy  reduces  relapses  in  the  radiation  portal 

and,  with  this  local  tumor  control,  allows  better  long-term  survival 

for  limited  disease  patients. 

113 

Cohen  notes  that  the  true  test  of  adjuvant  RT  is  whether  or 
not  it  increases  the  number  of  long-term  survivors — i.e.,  patients 
living  at  least  three  years — in  randomized  trials  comparing  RT  +  CT  to 
CT  alone. 

The  use  of  adjuvant  RT  in  extensive  disease  is  not  as 

controversial  a  topic.  Most  investigators  seem  to  agree  that  survival 

is  not  increased  by  RT  to  the  primary  tumor.  The  data  supporting  this 

114 

notion  are  relatively  scanty,  but  meticulous  local  control 


apparently  strikes  most  investigators  as  less  essential  when  the  tumor 
has  already  spread  beyond  one  hemithorax.  What  can  be  said  about 
local  control  and  its  relationship  to  long-term  survival? 


42 


Peschel  et  in  a  retrospective  review  of  12  patients 

achieving  survival  of  more  than  2  years,  stressed  the  need  for  local 

tumor  control — surgery  or  high  dose  (>4800  rads)  lung  irradiation — to 

avoid  local  relapse.  Three  of  5  patients  who  had  received 

chemotherapy  alone  or  low  dose  irradiation  (<3500  rads)  had  late  local 

83 

relapses.  Similarly,  Matthews  et  al.  reported  on  the  treatment 
received  by  patients  in  their  long  term  (>2.5  year)  survival 
registry.  The  two  largest  groups  represented  were  patients  who  had 
received  RT  +  CT  and  those  who  had  received  surgery  alone.  (The  role 
of  adjuvant  surgery  in  current  treatment  protocols  will  be  discussed 
later. ) 

Several  controlled,  randomized  studies  have  compared  CT  +  RT  to 

116 

CT  alone.  Hansen  et  al.  reported  shorter  median  survival  in  the  RT 

+  CT  group  compared  to  the  CT  group.  In  contrast,  Bunn  et  al.^^^  and 
1 18 

Perez  et  al.  reported  better  median  survival  and  complete  response 

rate  with  thoracic  irradiation.  The  Perez  study  also  reported  an 

initial,  significant  superiority  in  actuarial  3  year  survival  for  the 

group  receiving  RT  (20%  vs.  5%).  Toxicity  was  greater  in  the  RT  +  CT 

group.  There  were  2  induction  deaths  in  the  RT  +  CT  group  vs.  none  in 

119 

the  CT  group  in  Bunn's  study.  Mira  et  al.  have  added  RT  to  CT  at 
day  85  of  their  protocol  and  found  that,  in  about  1/3  of  responders 
who  did  not  achieve  complete  response  after  initial  CT,  RT  increased 
complete  response  rate  and  median  survival. 

Radiation  therapy  to  the  chest  has  a  logical  place  in  the  care  of 
patients  with  limited-stage  disease.  Local  control  is  an  extremely 


43 


useful  concept  in  designing  treatment  protocols  for  long-term 
survival.  Still,  the  trade-off  is  increased  toxicity. 

This  is  a  good  point  to  review  treatment  toxicities  briefly,  with 
a  special  interest  in  toxicities  associated  with  combined  modality 

120  12X3. 

therapy.  ’  Most  chemotherapy  regimens  used  for  treating  SCCL 

produce  some  degree  of  myelosuppression .  Addition  of  radiation 

affects  the  bone  marrow  and  in  a  healthy  adult,  ribs,  sternum,  and 

121b 

scapula  comprise  15-20%  of  functioning  bone  marrow.  With  most 

standard  CT  protocols  the  duration  of  granulocytopenia  is  relatively 

short;  febrile  episodes  are  reported  in  about  30%  of  patients, 

documented  infections  in  5%,  fatal  infections  in  2%.  When  adjuvant  RT 

is  added,  infections  have  been  reported  to  rise  to  11.7%,  fatal 

120 

infections  to  2.7%.  Infection  can  be  documented  in  about  40%  of 

febrile,  neutropenic  patients;  50%  of  these  have  bacteremia.  A  total 

of  60%  of  febrile,  neutropenic  patients  are  thought  to  be  infected  on 

120 

the  basis  of  cultures  or  clinical  signs  or  symptoms.  Thus, 

antimicrobial  therapy  is  empirically  employed  in  all  such  patients. 

Radiation  therapy  alone — but  especially  in  combination  with 
chemotherapy — contributes  to  two  major  acute  toxicities:  esophagitis 
and  pneumonitis. 

As  has  been  mentioned,  Adriamycin  enhances  radiation  induced 
esophagitis.  Chronic  esophageal  stricture  is  a  hazard  avoided  through 
careful  planning  of  the  portals  and  timing  of  RT  and  of  the  dose  and 
type  of  cytotoxic  therapy. 


1 


44 


CT  adds  to  the  problem  of  radiation  pneumonitis;  also,  chronic 
pulmonary  fibrosis  has  emerged  as  a  major  concern  in  long-term 
survivors  after  multi-modal  therapy. 

Cardiac  toxicity  is  a  potential  complication  of  SCCL  treatment. 
Pericarditis,  aggravation  of  coronary  artery  disease,  and 
cardiomyopathies  especially  associated  with  Adriamycin  are  all 
potential  toxicities. 

Peripheral  neuropathy  is  a  toxicity  associated  with  vincristine. 

The  long-term  neurological  sequelae  of  CT  +  RT  have  already  been 

discussed  in  the  context  of  prophylactic  cranial  irradiation. 

Finally,  second  malignancies  are  arising  as  toxic  complications. 

Four  cases  of  acute  leukemia — all  arising  2-1/2  to  3  years  after 

120 

diagnosis  of  SCCL — have  been  reviewed  by  Abeloff  et  al.  All  four 

patients  had  achieved  complete  responses;  3  of  the  4  had  received 
multi-modal  CT  +  RT  therapy. 

Adjuvant  surgery  is  a  final  topic  to  discuss  in  the  treatment  of 

limited-stage  SCCL.  Two  studies  have  been  mentioned  which  examined 

83  115 

the  characteristics  of  long-term  survivors  with  SCCL;  ’  in  each 
study,  patients  who  had  received  surgery  as  initial  or  only  treatment 
formed  a  significant  subgroup. 

122 

The  role  of  adjuvant  surgery  remains  unclear.  Comis  et  al. 

contributed  a  relatively  early  study,  which  they  have  recently 
123 

updated.  TNM  staging  was  used  for  the  surgical  procedure;  the 

authors  found  that  patients  with  superior  mediastinal  (N2)  disease  did 

124 

not  seem  to  benefit  from  adjuvant  surgery.  Foster  et  al. 


found 


45 


that — due  to  extent  of  disease  or  such  factors  as  poor  medical 

condition  and  inadequate  pulmonary  function — only  10  of  37  eligible 

125 

limited  disease  patients  were  surgical  candidates.  Friess  et  al., 

in  a  retrospective  review,  found  that  the  15  patients  with  limited 

disease  who  had  entered  one  of  their  combined  modality  protocols  after 

surgical  resection  had  significantly  better  median  and  2  year  survival 

than  patients  without  initial  surgery.  The  best  median  survival  was 

in  patients  with  the  smallest  lesions  (<5  cm)  who  had  undergone 

surgery  before  starting  the  protocol. 

Adjuvant  surgery  in  SCCL  may  become  an  accepted  treatment 

122 

modality.  Comis  et  al.  have  some  good  initial  results,  but  the 
number  of  patients  is  very  small.  Basic  questions  remain.  When  is 
adjuvant  surgery  most  effective?  (I.e.,  should  it  be  employed  before 
or  after  initial  chemotherapy?)  Is  adjuvant  surgery  only  possible  or 
efficacious  in  a  relatively  small  number  of  patients?  Finally:  are 
the  results  of  adjuvant  surgery  going  to  reflect  better  treatment  or 
simply  the  better  prognosis  of  a  subgroup  of  patients  with  "very 
limited"  stage  disease?^^ 


Extensive-Stage  Disease  and  Experimental  Therapies 
1 14 

Comis,  in  his  review  of  treatment  for  SCCL,  considers 
infrequent  long-term  survival  to  be  the  distinguishing  characteristic 
of  extensive-stage  disease.  Intensive  therapies  (high  dose,  high 
toxicity;  multiple,  novel  combinations;  new  drugs)  have  achieved 
better  median  survival.  A  glance  at  the  registry  of  long-term 


46 


QO 

survivors  (>2.5  years)  reported  in  1980  reveals  that,  of  97 
patients,  only  8  presented  with  extensive-stage  disease.  Extent  of 
disease  is  a  powerful  prognostic  indicator  and  survival  data  reflect 
this  fact. 

114 

Corais  cites  the  following  as  the  most  prevalent  new  approaches 
to  extensive  disease:  increasing  the  intensity  of  chemotherapy;  using 
a  sequence  of  drug  combinations  which  are  thought  to  be 
non-cross-resistant;  and  incorporating  Etoposide  (VP-16-213)  into  the 
initial  combination  of  drugs. 

Intensive  chemotherapy  seeks  to  take  advantage  of  dose-response 

relationships  and  of  the  intuitive  notion  that  if  "effective"  is  good 

65 

"intensive"  is  better.  Aisner  et  al.  point  to  the  paucity  of  data 

on  dose  schedule  dependency.  The  determination  of  maximum  doses 

proceeds  slowly,  on  a  drug-by-drug  basis.  Maximum  acceptable  toxicity 

appears  to  be  the  end-point.  The  results  have  not  been  encouraging 

and  toxicity  risks  are  considerable.  Late  intensive  combined  modality 

126 

therapy  with  autologous  bone  marrow  infusion  and  high-dose  therapy 

with  protected  environment-prophylactic  antibiotic  units  to  reduce 

127 

infectious  morbidity  have  been  reported  to  yield  no  long-term 
survival  advantage  over  more  conventional  therapy.  Neutropenia  and 
infection  are  prominent  risks.  High  dose  regimens  may  be  especially 
beneficial  in  patients  achieving  complete  response;^^’ however,  the 
generally  low  rate  of  complete  response  among  extensive  disease 
patients  limits  their  potential  application. 


47 


Another  novel  approach  to  therapy  is  the  use  of 

non-cross-resistant  drug  combinations  in  cycles.  The  results  have  not 
114  129 

been  exciting;  ’  still,  the  approach  may  hold  some  promise. 

130 

Evans  et  al.  have  pointed  out  that  most  alternating  sequences  of 
drugs  do  not  appear  to  be  truly  non-cross-resistant.  They  cite  a 


truly  non-cross  resistant  sequence  study  in  which  response  was 
131 

improved.  Still,  "truly  non-cross-resistant"  seems  to  mean  that 

potentially  better  response  is  achieved  by  achieving  potentially 

better  response — a  suspiciously  circular  chain  of  reasoning. 

New  drug  development  is,  of  course,  a  major  focus  of  continuing 

research.  These  drugs,  for  ethical  reasons,  are  usually  tested 

initially  in  patients  for  whom  first-line  chemotherapy  has  failed. 

Aisner  et  al.^^  note  the  hazards  of  this  approach.  It  may  be  that 

aggressive  initial  therapy  alters  the  nature  of  the  tumor  so  that  it 

becomes  refractory  to  any  subsequent  treatment.  (Evidence  that 

therapy  changes  biochemical  and  histological  characteristics  of  SCCL 

81  82 

tumors  has  already  been  noted  in  this  paper.  ’  )  Aisner  cites 

Etoposide  and  vindesine  as  examples.  Etoposide  is  probably  the  most 

active  single  agent  in  untreated  SCCL,  with  response  rates  averaging 
130 

over  40%.  Yet,  the  drug  has  generally  been  found  to  produce 

insignificant  response  rates  in  patients  refractory  to  standard 
130 

therapy.  Perhaps  the  problem  is  not  pre-treatment,  but  simply  that 

tumors  unresponsive  to  first-line  therapy  are  refractory  to  most  novel 
therapies  as  well. 


48 


In  any  case,  Etoposide  (VP-16-213)  has  proved  to  be  a  promising 

new  agent  in  treating  SCCL.  It  appears  to  show  a  dose-response 

relationship;  a  study  of  high-dose  Etoposide  achieved  an  80%  response 

132 

rate  in  10  patients  with  extensive  disease.  Etoposide  is  often 

used  in  current  multi-agent  chemotherapy  combinations. 

133  13^  133 

VM-26  (related  to  vincristine),  vindesine,  ’  and, 

136 

"logically,”  vindesine  +  Etoposide  may  have  activity  against 

SCCL.  The  latter  seems  a  good  example  of  combining  two  drugs  to  see 

if  the  combination  proves  to  have  some  magical  synergism.  Sometimes 

synergism  is  found.  When  Etoposide  alone  was  compared  to  Etoposide  + 

cis-platin  (EP)  in  patients  refractory  to  cyclophosphamide- 

130 

Adriamycin — vincristine  (CAV)  therapy  ,  the  EP  group  experienced  a 

better  response  rate,  higher  median  survival  and  increased 

thrombocytopenia  all  thought  to  reflect  the  synergistic  action  of 

Etoposide  and  cis-platin  described  in  some  animal  tumor 
137 

systems.  Since  their  patients  had  been  refractory  to  CAV  therapy, 

the  authors  suggested  they  may  have  found  a  truly  non-cross-resistant 

sequence  for  further  investigation  (CAV-EP).  The  usefulness  of  EP  as 

consolidation  therapy  after  initial  CAV  or  "combined  alkylators"  has 

been  reported  to  show  little  promise. 

Finally,  mention  should  be  made  of  two  studies  similar  to  the 

Yale  treatment  protocol  for  SCCL  whose  results  appear  in  the  next 

139 

section  of  this  paper.  Zekan  et  al.  found  that  CAVE  afforded 

significantly  increased  total  treatment  response  over  CAV  (82% 

vs.  66%).  Etoposide  was  said  to  have  added  little  toxicity  although 


49 


3/57  CAVE  patients  suffered  treatment-related  deaths  vs.  1/59  CAV 

patients.  Estimated  median  survival  was  not  significantly  different 

for  the  two  treatment  groups  in  limited  disease  or  extensive  disease. 

140 

Messeih  et  al.  reported  a  significantly  increased  overall 
response  rate  (65%  vs.  50%),  and  complete  response  rate  (44%  vs.  18%) 
for  their  CAVE  group  and — strikingly — extensive  disease  patients 
achieved  a  complete  response  rate  of  35%  on  CAVE  versus  0%  on  CAV. 
Overall  median  survival  for  all  responders  and  median  survival  for 
complete  responders  was  not  significantly  different  for  the  two 
treatment  groups. 


Closing  Comments 

Despite  the  tantalizing  response  of  SCCL  to  initial  radiation  or 
chemotherapy,  relapse  is  the  rule.  Long-term  survival  (best  defined 
as  longer  than  4-5  years  if  any  association  with  cure  is  to  be 
implied)  is  rare.  Extensive  disease  patients  have  an  especially 
dismal  prognosis  but  this  may  improve  if  more  can  achieve  complete 
response  to  therapy.  Still,  the  disease  remains  one  in  which  many 
patients  are  treated  to  allow  survival  of  a  few.  Severe  treatment 
toxicities  can  be  avoided  with  rational  dosage,  timing,  and  selection 
of  therapeutic  modalities.  They  should  be  avoided,  for  there  is  no 
evidence  that  toxic  therapies  are  the  best  therapies,  and  when  cure  is 
rare  treatment  should  be  relatively  palatable. 

Limited-stage  disease  offers  the  most  hope.  Prophylactic  cranial 
irradiation  (PCI)  appears  to  have  enough  chronic  neurological 


50 


toxicities  that  its  use  is  best  limited  to  complete  responders,  two  to 
four  months  after  the  start  of  therapy.  Thus,  PCI  will  be  employed 
mostly  in  limited-stage  disease.  PCI  may  fall  out  of  favor  entirely 
if,  for  example,  its  chronic  toxicities  are  found  to  outweigh  its 
protection  of  the  subgroup  of  patients  who  would  otherwise  suffer 
isolated  CNS  relapse.  Nitrosureas  appear  to  be  especially  associated 
with  the  chronic  toxicity  of  PCI.  Patients  with  less  than  complete 
responses  can  be  treated  for  CNS  metastases  as  they  arise.  Chest 
irradiation  makes  a  great  deal  of  sense  in  limited  disease;  there  is 
enough  clinical  evidence  and  good  theoretical  speculation  to  support 
the  notion  that  local  control  of  intrathoracic  disease  is  essential 
for  long-term  survival.  Care  must  be  taken  to  avoid  acute  toxicities 
that  accompany  multi-modal  therapy;  chronic  pulmonary  toxicity  is  a 
significant  factor  which  requires  further  study. 

The  importance  of  local  control  makes  adjuvant  surgery  a 
potentially  useful  treatment  modality.  The  apparently  superior 
survival  of  patients  with  small  '*very  limited"  tumors  highlights  the 
need  for  a  biochemical  marker  or  other  method  of  early  diagnosis 
before  SCCL  becomes  clinically  apparent.  If  high  risk  populations 
could  be  screened  for  the  disease,  survival  in  SCCL  would  certainly 
improve,  even  with  the  limitations  of  current  therapy. 

Our  understanding  of  SCCL  is  poor.  The  variability  of  treatment 
results  and  the  resistance  of  small  cell  tumors  to  second-line  drugs 
are  but  two  reflections  of  our  ignorance  in  the  clinical  setting.  The 
variability  among  pathologists  in  identifying  tumor  subtypes  and  the 


51 


lack  of  apparent  prognostic  significance  of  these  subtypes  make  the 
current  system  of  histological  classification  questionable. 

Heterogeneity  is  the  hallmark  of  SCCL;  tumor  cells  are  variable 
in  chromosome  number,  proliferative  activity,  antigenic  expression, 
clonal  origin,  cytomorphology  and  biochemical  behavior  (including 
expression  of  tumor  products  and  biochemical  markers).  Tumor  cells 
with  few  or  no  neurosecretory  granules,  low  dopa  decarboxylase  and 
bombesin  activity,  high  ploidy  and  active  proliferative  behavior  have 
all  been  identified  as  belonging  to  a  clinically  more  aggressive 
subclasss.  The  "variant"  subclass  of  tumor  cells  may  be  both 
radioresistant  and  more  aggressive  than  the  "classic"  subclass.  The 
origin  of  aggressive  tumor  cells  is  obscure  since  dopa  decarboxylase 
and  neurosecretory  granules  are  distinguishing  APUD  characteristics. 
Perhaps  they  evolve  from  cells  in  the  original  tumor  (i.e.,  the  tumor 
formed  by  initial  malignant  transformation). 

The  reclassification  of  SCCL  proposed  by  the  pathology  panel  of 
the  International  Association  for  the  Study  of  Lung  Cancer  is  of  great 
significance.  It  is  thought  that  the  NCI’s  "variant"  subclass  tumor 
cells  are  the  iui  vitro  equivalent  of  the  proposed  small  cell-large 
cell  subgroup,  and  that  the  NCI's  "classic"  cells  are  the  in  vitro 
equivalent  of  the  proposed  classic  small  cell  subgroup.  If,  for  the 
first  time,  a  prognostically  significant  classification  system  has 
been  found,  whose  subtypes  can  be  reliably  identified  by  different 
pathologists  and  studied  with  equivalent  in  vitro  cell  lines,  then  a 


major  step  will  have  been  taken  in  the  struggle  to  link  basic  science 


52 


research  on  cellular  characteristics  with  clinical  practice.  Until 
then,  information  on  the  heterogeneity  of  SCCL  tumor  cells  and  the 
cellular  characteristics  of  clinically  aggressive  tumors  goes  beyond 
our  ability  to  use  it:  the  information  doesn't  help  in  diagnosis,  for 
our  diagnostic  tools  detect  only  gross  disease;  it  doesn't  clarify  our 
histological  classification  system,  which  is  based  on  light 
microscopy;  it  doesn't  assist  us  in  prognosis,  which  is  based  on  gross 
extent  of  disease  and  subjective  evaluation  of  a  patient's  ability  to 
carry  out  his  daily  tasks;  and  it  probably  won't  help  us  design  better 
therapy,  since  our  therapeutic  modalities  are  so  very  limited.  But 
only  work  on  cells  will  characterize  the  SCCL  tumor.  Our  methods  of 
diagnosis,  classification,  prognosis,  and  treatment  will  become  more 
refined  as  understanding  of  the  tumor  cells  expands.  New 
modalities — hyperthermia,  monoclonal  antibodies,  radiosensitizing 
drugs^ — may  prove  useful  by  empirical  trial.  Today's  dilemma  is  that 
a  hit-or-miss  approach  to  SCCL  is  bound  to  fail  and  the  information  we 
need  for  rational  therapy  is  elusively  basic. 


1 

'1 


53 


PART  THREE:  THE  STUDY 

This  paper  presents  the  initial  results  of  a  Yale  University 
treatment  protocol  for  small  cell  cancer  of  the  lung  (SCCL).  The  data 
are  part  of  a  continuing  study;  methods,  patients,  results,  and 
discussion  are  presented  below. 


Methods 

During  the  period  October,  1980  to  April,  1983  all  referred 
patients  with  histologically  confirmed  SCCL  (by  cytology  or  biopsy  of 
metastatic  sites)  were  entered  in  the  study.  Patients  were  accepted 
regardless  of  stage  of  disease,  performance  status,  or  life 
expectancy,  provided  they  had  at  least  one  site  of  measureable  or 
evaluable  disease.  Patients  were  ineligible  for  inclusion  in  the 
study  if  they  had  received  prior  treatment  for  their  disease,  with  the 
exception  of  surgery,  or  if  their  left  ventricular  ejection  fraction, 
by  gated  blood  pool  scan,  was  too  low  to  permit  treatment  with 
Adriamycin  (doxorubicin). 

Pretreatment  staging  evaluation  included  history  and  physical 
examination  with  evaluation  of  performance  status.  Blood  tests 
included  CBC,  platelet  count,  BUN,  creatinine,  bilirubin 
(total/direct),  alkaline  phosphatase,  glucose,  electrolytes, 
prothrombin  time/partial  thromboplastin  time,  cortisol,  and  studies 
for  ectopic  hormones  as  indicated. 


54 


Diagnostic  procedures  included  bone  marrow  biopsy  and  aspirate; 
chest  x-ray  with  tomography  in  all  patients  with  limited-stage  disease 
and  others  as  indicated;  liver-spleen  scan  and  bone  radionuclide 
scans;  CT  scan  of  the  head;  skin  tests  for  SKSD,  PPD,  Candida,  mumps; 
electrocardiogram;  and  left  ventricular  ejection  fraction  gated  blood 
pool  scan. 

Patients  were  defined  as  having  limited-stage  disease  (LD)  if  the 
disease  was  confined  to  one  hemithorax,  with  or  without  involvement  of 
hilar,  mediastinal  and  ipsilateral  supraclavicular  lymph  nodes. 
Extensive-stage  disease  (ED)  was  defined  as  disease  beyond  these 
confines. 

For  treatment,  patients  were  randomized  prospectively  to  CAV 

(cyclophosphamide,  Adriamycin,  vincristine)  or  CAV/E  (the  above  plus 

2 

Etoposide  (VP-16-213).  Drug  dosages  were:  Adriamycin  40  mg/m  ; 

2  2 

cyclophosphamide  1000  mg/m  IV;  vincristine  1.4  mg/m  IV  (not  to 

2 

exceed  a  total  dose  of  2  mg);  Etoposide  125  mg/m  IV.  CAV  cycles  were 

every  21  days.  CAV/E  cycles  were  every  42  days,  with  CAV  given  on  day 

2 

1,  Etoposide  125  mg/m  IV  on  each  of  days  21,  23,  and  25,  beginning 
again  with  CAV  on  day  42. 

In  limited  disease,  3000  rads  of  radiation  therapy  (RT)  to  the 
primary  tumor,  mediastinum,  and  bilateral  supraclavicular  nodes  as  300 
rads  per  day,  5  treatments  per  week  (10  treatments  total)  was  given 
initially.  Vincristine  and  cyclophosphamide  in  the  doses  above  were 
given  after  staging,  concurrent  with  the  first  phase  of  RT,  followed 
by  4  cycles  of  CAV  or  2  cycles  of  CAV/E.  Adriamycin-containing 


I 


;■  t 


^ni  l.'i 


55 


combination  therapy  thus  began  after  completion  of  the  first  phase  of 
RT  and  at  least  21  days  after  initial  cyclophosphamide  and 
vincristine.  An  additional  2400  rads  of  RT  to  the  primary  sites,  with 
concurrent  cyclophosphamide  and  vincristine,  were  given  as  8 
treatments  of  300  rads  each,  after  the  first  4  cycles  of  CAV  or  2 
cycles  of  CAV/E,  before  completing  6  more  cycles  of  CAV  or  3  more 
cycles  of  CAV/E. 

In  extensive  disease,  treatment  was  as  above,  except  irradiation 
of  the  primary  site  was  at  the  option  of  the  responsible  clinician. 

After  cycle  4  of  CAV  or  cycle  2  of  CAV/E,  all  complete  responders 
with  no  known  brain  metastases  received  prophylactic  whole  brain 
irradiation  as  3000  rads  over  2  weeks  at  300  rads  per  treatment, 
regardless  of  disease  extent  at  presentation. 

Treatment  was  continued  to  10  cycles  of  CAV  or  5  cycles  of  CAV/E. 

See  Tables  9  and  10  for  summaries  of  the  treatment  protocols. 

If,  after  6-8  weeks  of  chemotherapy,  there  was  disease 
progression,  patients  were  considered  off-study  and  treatment  was 
individualized.  Otherwise,  patients  were  treated  per  protocol  until 
clear-cut  evidence  of  progression  or  relapse.  Subsequent  therapy  was 
individualized. 

At  the  conclusion  of  therapy,  patients  were  restaged  to  document 
response. 

Dose  attenuations  were  guided  by  CBC  prior  to  therapy. 

Complete  response  was  defined  as  total  disappearance  of  all 
disease  with  biopsy  confirmation  (e.g.,  for  bone  marrow  or  liver) 


56 


lasting  at  least  30  days. 

Partial  response  was  defined  as  a  50%  decrease  in  the  product  of 
2  tumor  diameters  perpendicular  to  one  another,  without  associated 
progression  of  any  other  lesions  or  the  appearance  of  a  new  lesion. 
Regression  had  to  last  a  minimum  of  60  days. 

Stable  disease  was  defined  as  less  than  50%  regression  of 
measureable  lesions  with  the  appearance  of  no  new  lesions  and  no 
deterioration  of  performance  status. 

Progression  of  disease  was  defined  as  the  appearance  of  any  new 
lesion  or  the  increase  in  size  of  any  measureable  lesion  by  greater 
than  50%. 

In  this  report,  patients  with  stable  disease  and  progressive 
disease  are  grouped  together  as  "non-responders." 

ECOG  toxicity  criteria  were  used  as  a  basis  for  patient 
comparison. 

Performance  status  was  defined  as  follows:  0-asymptomatic ; 
1-fully  ambulatory  with  symptoms;  2-bedridden  less  than  50%  of  the 
time;  3-bedridden  50%  of  the  time  or  more;  4-100%  bedridden. 

Statistical  analysis  of  time  to  relapse  and  survival  was 
performed  using  Kaplan-Meier  plots;  comparisons  were  made  using  the 
generalized  Wilcoxon  (Breslow)  test  of  statistics.  All  median  values 
are  from  the  Kaplan-Meier  plots  and  therefore  may  be  projections. 


57 


Patients  and  Results 

Of  the  47  patients  entered  into  the  study,  8  were  inevaluable. 
Five  of  the  8  patients  had  extensive  disease  (ED).  Of  these  5:  2 
patients  never  got  Adriamycin  due  to  inadequate  pre-treatment  cardiac 
function;  1  had  intercurrent  prostatic  cancer;  1  chose  to  leave  the 
care  of  a  physician  participating  in  the  study  after  one  visit,  for 
unknown  reasons;  and  1  patient  had  a  sudden  cardiac  death  48  hours 
after  her  only  cycle  of  CAV  therapy. 

Three  of  the  8  inevaluable  patients  had  limited  disease  (LD). 

Of  these  3:  1  patient  had  not  been  on-study  long  enough  to  evaluate 

response — in  addition,  this  patient's  tumor  was  of  mixed  small 
cell/large  cell  histology;  1  had  intercurrent  prostate  cancer;  and  1 
patient's  chemotherapy  was  discontinued  at  the  patient's  request  when 
symptoms  of  congestive  heart  failure  developed  after  one  dose  each  of 
cyclophosphamide  and  vincristine  (the  cycle  contained  no  Adriamycin). 

On-study  time  was  defined  as  the  date  treatment  started  to  the 
date  last  seen  or  date  of  death.  The  39  evaluable  patients  had  a 
median  on-study  time  of  219  days  (range  6-907  days). 

Twenty-eight  of  39  patients  have  relapsed.  Eleven  of  39  patients 
have  not  relapsed,  one  of  whom  died  without  apparent  relapse  (of 
infection  or  radiation  pneumonitis,  as  will  be  described  later);  the 
other  ten  patients  are  still  living  and  are  disease-free.  Sixteen  of 
39  patients  are  still  alive,  including  6  who  have  relapsed.  The 
median  follow-up  for  patients  still  alive  is  219  days  (two  shortest 


58 


follow-ups:  70  and  97  days;  two  longest:  674  and  907  days). 

Patient  characteristics  are  presented  in  Table  11,  subdivided  by 
extent  of  disease  and  treatment  arm.  Fifteen  of  39  patients  (38%)  had 
limited  disease  (LD).  Twenty-four  of  39  (62%)  had  extensive  disease 
(ED).  Two  ED  patients  presented  with  superior  vena  cava  syndrome;  1  ED 
patient  had  SIADH  on  presentation.  One  LD  patient  had  significant 
non-neoplastic  disease  on  presentation  (diffuse  scleroderma;  she  is 
the  only  patient  whose  initial  performance  status  is  unknown). 

As  expected,  LD  patients  had  better  initial  performance  status 
than  ED  patients  (LD — 11  of  15  patients  fully  ambulatory  (performance 
status  0  or  1);  ED — 11  of  24  patients  fully  ambulatory).  The  LD  group 
was  slightly  younger  than  the  ED  group  (median  ages:  LD-60  years; 
ED-63.5  years).  The  LD  group  contained  a  greater  proportion  of  women 
(LD-  9  women: 6  men;  ED-  11  women: 13  men). 

Comparing  treatment  arm  groups  (Table  11):  On  the  whole,  the  CAV 
group  contained  younger  patients  (median  ages:  CAV-59  years;  CAV/E-66 
years).  LD-ED  distribution  was  similar  for  both  treatment  groups:  of 
21  CAV  patients,  there  were  8  LD  (38%)  and  13  ED  (62%);  of  18  CAV/E 
patients,  there  were  7  LD  (39%),  11  ED  (61%). 

Fourteen  of  21  CAV  patients  (67%)  were  fully  ambulatory 
(performance  status  0  or  1)  versus  8  of  18  CAV/E  patients  (44%).  Most 
of  this  difference  can  be  accounted  for  by  the  fact  that  8  of  13 
patients  (62%)  in  the  ED-CAV  group  were  fully  ambulatory  versus  3  of 
11  patients  (27%)  in  the  ED-CAV/E  group. 


59 


The  CAV/E  group  contained  a  greater  proportion  of  women  (CAV/E- 
10  women: 8  men;  CAV-  10  women: 11  men). 

Three  patients  had  surgery  before  beginning  the  protocol:  2  LD, 

1  ED.  Patient  characteristics  are  continued  in  Tables  12  and  13. 

Eighteen  of  24  ED  patients  presented  with  metastatic  disease  in 
more  than  one  site.  Sites  of  presenting  metastases,  by  treatment  arm, 
with  the  number  and  percentage  of  patients  presenting  with  them  are 
shown  in  Table  12.  Six  of  24  ED  patients  presented  with  metastatic 
disease  involving  single  sites  (see  Table  12). 

Sites  of  relapse  among  all  39  patients  (ED  +  LD)  with  the  number 
and  percentage  of  patients  relapsing  at  those  sites  are  presented  in 
Table  13.  There  were  10  relapses  in  sites  of  initial  disease, 
excluding  the  chest  (see  Table  13). 

Of  the  5  brain  relapses,  4  occurred  in  ED  patients  who  had 
received  no  prophylactic  cranial  irradiation  (PCI).  Three  of  these  4 
patients  received  no  CT  scan  or  radionuclide  brain  scan  on  diagnosis. 
One  of  the  5  brain  relapses  occurred  in  an  LD  patient  with  negative  CT 
scan  on  diagnosis  who  relapsed  4  months  after  3000  rads  of  PCI.  The 
ED  patient  who  experienced  a  choroidal  relapse  had  no  PCI. 

Two  LD  patients  deserve  special  mention.  The  first  patient  had  a 
palpable  subcutaneous  nodule  at  diagnosis,  refused  biopsy,  and  later 
relapsed  in  the  same  site;  the  second  had  a  radionuclide  scan 
equivocal  for  liver  involvement  at  diagnosis  and  later  relapsed  in 
liver,  bone  and  bone  marrow. 


60 


Nine  patients  had  chest  relapses.  Five  of  the  9  had  ED  and 
received  no  thoracic  irradiation.  It  is  not  known  whether  the 
remaining  4  patients  (2  LD;  2  ED)  relapsed  within  their  radiation 
therapy  portals. 

Response  to  therapy,  grouped  by  disease  extent  and  treatment  arm, 
is  presented  in  Table  14.  Objective  responses  (CR  +  PR)  occurred  in  28 
of  all  39  patients  (72%);  in  13  of  15  LD  patients  (87%);  15  of  24  ED 
patients  (63%);  8  of  8  LD-CAV  patients  (100%);  5  of  7  LD-CAV/E 
patients  (71%);  9  of  13  ED-CAV  patients  (69%);  and  6  of  11  ED-CAV/E 
patients  (55%). 

The  following  sections  present  data  from  Kaplan-Meier  curves  for 
time  to  relapse  and  survival.  Subgrouping  was  performed  in  analyzing 
the  data  by  treatment  group  (e.g.,  LD-CAV  responders  vs.  LD-CAV/E 
responders);  such  subgrouping  is  intended  only  to  reflect  the 
distribution  of  the  data,  since  the  small  number  of  patients  in  these 
subgroups  precludes  in-depth  analysis. 

Time  to  relapse  was  defined  as  the  date  treatment  began  to  the 
date  of  disease  progression.  The  data  are  presented  in  Table  14  and 
Figures  1-4. 

Median  time  to  relapse  was  361  days  in  responders  (CR  +  PR)  with 
LD;  for  ED  responders,  the  median  was  188  days.  Analysis  of  these 
relapse  curves  showed  a  significantly  longer  time  to  relapse  for  the 
LD  responders  (£_=.0001).  (See  Figure  1.)  Time  to  relapse  for  ED 
non-responders  (median:  71  days)  was  significantly  shorter  than  for 
ED  responders  (£_=.006).  (See  Figure  2.) 


61 


Time  to  relapse  was  studied  by  treatment  group.  For  patients 
with  LD,  time  to  relapse  on  CAV  (median:  334  days)  versus  time  to 
relapse  on  CAV/E  (median:  361  days)  was  not  significant 
(£=•55).  (See  Figure  3.)  In  contrast,  time  to  relapse  for  all  ED 
patients  on  CAV  (median:  193  days)  compared  to  ED  patients  on  CAV/E 
(median:  109  days)  was  barely  significant  (£=.04).  (See  Figure 

4. )  Further  subgrouping  revealed  that  time  to  relapse  of  ED 
responders  (CR  +  PR)  on  CAV  versus  those  on  CAV/E  was  not  significant 
(£=.77);  but  comparison  of  ED  non-responders  on  CAV  versus  ED 
non-responders  on  CAV/E  was  significant  (£=.02). 

Survival  data,  the  main  criteria  by  which  protocols  are 
evaluated,  are  presented  in  Table  15  and  Figures  5-9. 

Median  survival  for  all  patients  was  301  days.  (See  Figure 

5. )  Median  survival  for  LD  complete  responders  was  560  days. 

Survival  of  LD  responders  (CR  +  PR)  (median:  560  days)  was 
compared  to  survival  of  ED  responders  (median:  230  days)  and  found  to 
be  significant  (£=.0007).  (See  Figure  6.)  Survival  of  ED  responders 
versus  ED  non-responders  (median:  198  days)  was  not  significant 
(£=.24).  (See  Figure  7.) 

Survival  by  treatment  group  was  analyzed.  When  survival  of  LD 
patients  on  CAV  (median:  560  days)  was  compared  to  LD  patients  on 
CAV/E  (median:  424  days),  no  significant  difference  was  found 
(£=.24).  (See  Figure  8.)  Survival  of  ED  patients  on  CAV 
(median:  230  days)  versus  ED  patients  on  CAV/E  (median:  186  days) 
was  not  significant  (£=.23).  (See  Figure  9.) 


62 


A  glance  at  the  survival  curve  for  all  patients  shows  a  plateau 
at  about  6%.  (See  Figure  5.)  The  curve  for  LD  complete  responders 
plateaus  at  35%.  But  8  of  the  12  patients  in  this  group  were  still 
alive  and  those  8  had  a  median  follow-up  of  only  305  days,  while 
projected  median  survival  was  560  days. 

Thusfar,  4  patients  have  lived  1-1/2  years  or  more.  Two  are 
described  in  some  detail  below  because  they  will  be  mentioned  in  the 
discussion  of  treatment  results  later  on. 

First,  a  male  patient  presented  at  51  years  of  age  with 
performance  status  1  and  extensive  disease — bone  involvement,  pleural 
effusion  of  unknown  cytology,  a  subcutaneous  nodule  in  the  left  flank 
and  a  supra-clavicular  node.  After  4  cycles  of  therapy,  his  chest 
disease  had  not  changed  significantly;  however,  he  experienced  a 
choroidal  relapse  in  the  left  eye,  with  detachment  and  uplifting  of 
the  retina.  The  patient  received  radiation  therapy  to  the  eye  and 
additional  cycles  of  CAV.  His  chest  x-ray  shov;ed  no  significant 
improvement  during  7  months  of  therapy — thus,  he  was  a  non-responder. 
However,  he  did  not  expire  until  847  days  after  the  start  of  therapy. 

Second,  a  60-year-old  woman  with  limited  disease  and  performance 
status  0  underwent  a  left  lower  lobectomy  then  received  CAV  therapy, 
achieving  complete  response.  She  was  still  alive  at  907  days,  without 
relapse. 

Some  toxicities  were  common  but  not  severe  enough  to  cause  great 
concern:  radiation  esophagitis  (never  causing  strictures  or  requiring 

hospitalization);  nausea  and  vomiting  (controllable);  mucositis  (never 


63 


precluding  oral  food  consumption);  alopecia.  All  were  ECOG  #2 
(moderate  toxicity)  or  better. 

Myelosuppression  significant  enough  to  cause  a  drop  in  WBC  count 
to  <2000  (ECOG  #3  or  worse)  was  experienced  by  24  of  39  patients 
(62%):  14  of  24  patients  with  ED  (58%)  and  10  of  15  with  LD  (67%);  by 
treatment  group:  16  of  21  CAV  patients  (76%);  8  of  18  CAV/E  patients 
(44%). 

Anemia  severe  enough  to  require  transfusion  (ECOG  #3)  was 
experienced  by  9  of  39  patients  overall  (23%):  5  of  24  with  ED  (21%); 
4  of  15  with  LD  (27%);  6  of  21  on  CAV  (29%);  3  of  18  on  CAV/E 
(17%).  Three  patients  require  special  mention:  one  extensive  disease 
patient  on  CAV  had  a  Hgb/Hct  of  8.6/25.3  but  no  transfusion 
documented;  one  LD-CAV/E  patient  had  Hgb/Hct  of  10.1/25.5  but  refused 
transfusion;  one  ED-CAV  patient  had  chronic  anemia  status  post  Bilroth 
II  surgery  and  his  anemia  was  not  evaluable  as  a  toxicity. 

No  platelet  counts  <50,000  (ECOG  #3  or  worse)  were  documented  and 
there  were  no  episodes  of  bleeding. 

Six  patients  were  hospitalized  8  times  for  pneumonia;  3  episodes 
of  concurrent  sepsis  were  documented.  Two  patients  were  hospitalized 
three  times  for  fever:  one  of  these  patients  was  hospitalized 
separately  for  pneumonia,  and  is  included  among  such  patients  above; 
one  patient  was  hospitalized  twice  with  negative  cultures  but  a  left 
upper  lobe  cavitary  lesion  on  chest  x-ray  and  a  positive  PPD  test. 

The  latter  patient  was  treated  with  INH  and  Rifampin. 


64 


One  patient  was  hospitalized  once  with  a  lung  abcess  and  failure 
to  thrive. 

One  patient  was  hospitalized  once  for  pancytopenia  (WBC  count  of 
300)  but  neither  fever  nor  infection  was  documented. 

Eight  patients  were  thus  considered  to  have  been  hospitalized  at 
some  time  for  infection  (all  those  described  above  except  the  patient 
with  pancytopenia  only).  Seven  of  these  8  patients  had  ED;  the  one  LD 
patient  was  hospitalized  twice,  once  for  pneumonia  without  sepsis, 
once  for  fever  only. 

Thus,  7  of  24  ED  patients  (29%)  experienced  significant  infection 
as  did  1  of  15  LD  patients  (7%);  7  of  21  CAV  patients  (33%);  and  1  of 
18  CAV/E  patients  (6%).  One  patient  not  included  above  may  have  died 
of  treatment-related  infection,  as  discussed  below. 

Two  patients,  both  with  LD  on  CAV/E,  experienced  radiation 
pneumonitis,  one  requiring  treatment  with  steroids.  A  third  patient, 
with  ED  on  CAV,  who  had  superior  vena  cava  syndrome  and  liver 
involvement  at  presentation,  was  hospitalized  12  days  after  her  last 
chemotherapy  cycle,  5  weeks  after  radiation  therapy  to  the  chest,  with 
leukopenia,  fever,  chills,  and  bilateral  pulmonary  infiltrates. 
Cultures  were  negative,  but  she  was  begun  on  antibiotics.  Her  lung 
disease  was  thought  to  be  consistent  with  radiation  pneumonitis,  but 
this  was  diagnosed  by  chest  x-ray  and  clinical  impression  only.  The 
patient  progressed  to  "Adult  Respiratory  Distress  Syndrome"  after  one 
week  of  hospitalization  and  expired  two  and  one-half  weeks  after 
admission.  This  patient  almost  certainly  died  of  treatment-related 


\ 


65 


toxicity.  Infection  is  thought  to  be  the  most  likely  cause;  radiation 
pneumonitis  is  a  possibility.  The  patient  died  without  documented 
relapse  after  a  partial  response  to  therapy. 

One  inevaluable  patient  had  a  possible  treatment-related  death. 
She  was  a  70-year-old  woman  who  presented  with  performance  status  4, 
SIADH  and  extensive  disease.  Her  cardiac  ejection  fraction  was 
50%.  She  received  one  cycle  of  CAV  therapy  and  had  a  sudden  cardiac 
death  48  hours  later. 

Three  other  patient  deaths  should  be  described. 

A  patient  with  LD  on  CAV  therapy  who  received  prophylactic 
cranial  irradiation  (PCI)  developed  dementia,  dizziness,  and  double 
vision.  Her  CNS  symptoms  progressed,  and,  in  light  of  a  lumbar 
puncture  and  CT  scan  negative  for  tumor,  she  was  felt  to  have  died  of 
paraneoplastic  encephalopathy.  However,  combined  Adriamycin-radiation 
therapy  toxicity  cannot  be  ruled  out. 

One  patient  with  ED,  a  non-responder  to  CAV  therapy,  experienced 
dementia  with  memory  loss  and  confusion.  The  patient’s  CNS  symptoms 
progressed  and,  in  light  of  a  CT  scan  and  lumbar  puncture  negative  for 
tumor,  his  death  was  felt  to  be  consistent  with  paraneoplastic 
encephalopathy.  The  patient  received  no  PCI.  However,  death  due  to 
toxicity  of  chemotherapy  alone  cannot  be  ruled  out. 

Finally,  a  58-year-old  man  presented  with  significant  liver 
involvement,  bilateral  lymphadenopathy ,  and  performance  status  of 
3.  He  was  randomized  to  CAV/E  therapy  and  died  due  to  progression  of 
his  disease  in  the  liver  6  days  after  his  only  therapy  cycle,  which 


66 


consisted  of  CAV.  This  patient  is  mentioned  because  his  case  will  be 
noted  in  the  discussion  of  treatment  results  later  in  the  paper. 

Three  patients  experienced  significant  cutaneous  infections.  Two 
patients — one  LD  on  CAV/E,  one  ED  on  CAV — experienced  H.  simplex 
infections  while  being  hospitalized  for  concurrent  problems.  An  ED 
patient  on  CAV/E  experienced  an  H.  Zoster  infection  as  an  outpatient. 

Three  patients  had  rash  reactions  to  chemotherapy:  one  to 
Adriamycin;  one  to  Adriamycin  and  cyclophosphamide  or  cyclophosphamide 
alone;  one  unknown.  Two  patients  required  treatment  with  IV 
steroids. 

Adriamycin  had  to  be  discontinued  in  2  patients  due  to 
cardiotoxicity .  None  experienced  heart  failure  (both  toxicities  ECOG 
#2).  One  inevaluable  patient  experienced  heart  failure  after  a  single 
cycle  of  chemotherapy  which  did  not  contain  Adriamycin.  Chemotherapy 
was  discontinued  at  the  patient's  request. 

Vincristine  neurotoxicity  was  significant  enough  to  cause 
discontinuation  of  the  drug  in  4  patients  (3  LD  on  CAV;  1  ED  on 
CAV/E).  One  of  these  patients  (ED)  experienced  "Etoposide  accentuated 
vincristine  neuropathy  with  foot  drop”  and  both  drugs  were 
discontinued.  The  only  other  documented  attenuation  of  Etoposide  was 
one  cycle  of  3  doses  for  myelosuppression  just  before  the  patient 
relapsed.  Vincristine  dose  attenuation  of  more  than  50%  was  required 
in  4  patients  (2  ED-CAV;  1  ED-CAV/E;  1  LD-CAV)  for  whom 
discontinuation  of  the  drug  was  not  necessary. 


67 


Finally,  7  patients  required  significant  attenuations  (>50%)  in 
the  dose  of  their  chemotherapy  (cyclophosphamide  and/or  Adriamycin) 
for  myelosuppression  alone:  3  ED-CAV;  1  ED-CAV/E;  2  LD-CAV/E;  1 
LD-CAV. 


Discussion 

The  International  Association  for  the  Study  of  Lung  Cancer 
Workshop  has  published  its  treatment  result  expectations  for 
SCCL.^^  Expectations  include  complete  response  of  50%  in  LD  and  20% 
in  ED;  median  survival  of  at  least  14  months  in  LD  and  7  months  in  ED; 
and  15-20%  3  year  disease-free  survival  among  LD  patients.  These 
expectations  may  be  excessively  optimistic  (especially  those  for 
long-term  survival — see  Table  8),  but  at  least  they  establish  some 
standard  for  comparison.  Table  7  presents  treatment  results  from 
selected  studies;  direct  comparisons  are  not  possible  between 
studies. 

The  response  rates  in  the  present  series  were  generally  good, 
with  80%  of  LD  patients  achieving  complete  response.  Just  12%  of  ED 
patients  achieved  complete  response,  a  low  but  acceptable  number. 

Projected  median  survival  was  very  good,  ranging  from  about  6.5 
months  for  ED  non-responders  to  more  than  18  months  for  LD  complete 
responders.  As  expected,  both  time  to  relapse  and  survival  were 
significantly  longer  for  LD  responders  (CR  +  PR)  than  for  ED 
responders.  The  presence  in  the  ED  non-responder  group  of  a  patient 
who  survived  847  days  after  a  choroidal  relapse  must  be  kept  in  mind 


68 


when  evaluating  the  projected  survival  data  for  this  relatively  small 
group  (n=9).  The  unusual  choroidal  relapse  appears  to  have  had  no 
significant  negative  influence  on  this  patient's  survival.  The 
projected  median  survival  of  560  days  for  LD  complete  responders  must 
also  be  approached  with  some  caution  since  the  projection  is  based  on 
data  from  12  patients,  8  of  whom  were  still  living.  The  living 
patients  had  a  median  follow-up  of  just  305  days. 

Of  the  12  LD  patients  achieving  complete  response,  one  was  alive 
and  disease-free  at  907  days  (slightly  less  than  2.5  years). 
Interestingly,  this  patient  had  surgery  prior  to  beginning  the 
protocol,  adding  further  anecdotal  evidence  to  the  efficacy  of 
adjuvant  surgery  in  achieving  local  control  and  the  importance  of 
local  control  in  long-term  survival.  In  fact,  it  is  too  soon  to 
predict  the  number  of  long-term  survivors  from  this  study. 

The  two  treatment  groups  were  very  similar  in  survival  results. 
The  CAV/E  group  had  shorter  projected  median  survival  in  both  ED  and 
LD,  but  no  comparison  with  CAV  survival  curves  was  significant.  Time 
to  relapse  was  shorter  for  the  ED-CAV/E  group  than  the  ED-CAV  group 
and  the  comparison  was  barely  significant  (_p^=.04).  Further 
subgrouping  showed  a  significantly  shorter  time  to  relapse  for  the  ED 
non-responders  on  CAV/E  than  those  on  CAV.  Comparison  of  time  to 
relapse  for  ED  responders  was  not  significantly  different  for  the  two 
treatment  groups.  There  are  numerous  reasons  for  quicker  time  to 
relapse  in  ED  non-responders  on  CAV/E.  These  include  the  fact  that 
the  ED-CAV/E  group  contained  a  substantially  smaller  proportion  of 


69 


fully  ambulatory  patients  than  the  ED-CAV  group  (62%  vs.  27%).  The  ED 
non-responder  CAV  group  contained  that  patient  with  the  choroidal 
relapse  who  went  on  to  relatively  long  survival  while  the  ED-CAV/E 
non-responder  group  contained  the  patient  who  died  of  progressive 
disease  in  only  6  days.  ED  non-responders  on  CAV  survived  longer  than 
ED  non-responders  on  CAV/E  but  the  groups  are  small  and  comparison 
didn't  quite  reach  significance  (2.=  . 053).  Interestingly,  survival  was 
better  for  the  ED  responders  on  CAV/E  than  those  on  CAV,  but  the 
comparison  was  not  significant  (2=*'^1)* 

Clearly,  the  addition  of  Etoposide  to  CAV  produced  no  difference 

139,140 

in  treatment  results  worthy  of  mention.  iwo  previous  reports 
cited  better  response  rates  with  the  addition  of  Etoposide  to  CAV; 
still,  the  studies  found  no  significant  differences  between  the 
treatment  groups  in  survival. 

No  unexpected  toxicities  arose  in  the  study.  Myelosuppression 

was  no  greater  than  that  consonant  with  a  good  therapeutic  response. 

Etoposide  added  no  apparent  additional  toxicity  to  the  CAV 

regimen.  In  fact,  only  1  of  the  18  CAV/E  patients  (6%)  required 

hospitalization  for  infection  versus  7  of  the  21  CAV  patients 

(33%).  The  33%  rate  for  CAV  patients  is  higher  than  the  11.7% 

"standard"  infection  rate  for  combined  modality  protocols  cited  in  one 

120 


review  of  SCCL  treatment  complications. 


The  6%  rate  with  CAV/E  is 


lower  than  the  "standard"  rate  and  unexpected.  Leukopenia  (WBC  count 
<2000)  was  experienced  by  16  of  21  CAV  patients  (76%)  versus  8  of  18 
CAV/E  patients  (44%).  Perhaps  this  underlies  the  difference  in 


# 


70 


infection  rates,  unless  Etoposide  has  some  heretofore  undiscovered 
antibiotic  properties. 

One  treatment-related  death  was  probably  caused  by  infection 

(although  radiation  pneumonitis  is  possible),  yielding  a  fatal 

infection  rate  of  1  in  39  patients  (2.6%).  This  is  reasonable  for  a 

120 

combined  modality  study.  Sudden  cardiac  death  occurred  after  a 

single  cycle  of  cyclophosphamide  and  vincristine  in  a  patient 
presenting  with  extensive  disease,  SIADH,  poor  performance  status 
(bedridden)  and  a  cardiac  ejection  fraction  of  50%.  This  must  be 
viewed  as  a  possible  treatment-related  death  although  no  Adriamycin 
was  given. 

In  summary,  the  present  study  employed  state-of-the-art  design 

(prophylactic  cranial  irradiation  after  complete  response,  thoracic 

irradiation  in  limited  disease,  and  use  of  Etoposide,  an  agent  with 

significant  activity  against  SCCL)  and  achieved  early  treatment 

results  comparable  to  those  in  the  current  literature.  It  is  too 

early  to  evaluate  long-term  survival. 

Addition  of  Etoposide  to  CAV  therapy  yielded  no  improvement  in 

initial  treatment  results,  including  survival.  However,  an 

unexpectedly  low  rate  of  infections  requiring  hospitalization  was 

found  in  the  CAV/E  group,  substantially  lower  than  that  in  the  CAV 
141 

group,  perhaps  secondary  to  a  lower  rate  of  leukopenia  with  the  use 
of  Etoposide  in  half  the  treatment  cycles. 


71 


REFERENCES 

1.  Stolley,  P.D.:  Lung  cancer  in  women — five  years  later, 
situation  worse.  N.  Engl,  J.  Med.  309:428-429,  1983. 

2.  Silverberg,  E.:  Cancer  statistics,  1984.  CA  34:7-15,  1984. 

3.  Weiss,  W. :  Small  cell  carcinoma  of  the  lung:  Epidemiology  and 
etiology,  in  Greco,  F.A.,  Oldham,  R.K.,  Bunn,  P.A.,  Jr. 

(eds.):  Small  Cell  Lung  Cancer  (Clinical  Oncology 
Monographs).  New  York,  Grune  and  Stratton,  1981,  pp.  1-34. 

4.  Annegers,  J.F.,  Carr,  D.T.,  et  al.:  Incidence,  trend,  and 
outcome  of  bronchogenic  carcinoma,  in  Olmsted  County,  Minnesota, 
1935-1974.  Mayo  Clin.  Proc.  53:432-436,  1978. 

5.  Hoffman,  P.C.,  Albain,  K.S.,  et  al.;  Current  concepts  in  small 
cell  carcinoma  of  the  lung.  CA  34:269-281,  1984. 

6.  Boucot,  K.R.,  Weiss,  W.,  et  al.,:  The  Philadelphia  pulmonary 
neoplasm  research  project:  Basic  risk  factors  of  lung  cancer  in 
older  men.  American  Journal  of  Epidemiology  95:4-16,  1972. 

7.  Carney,  D.N.,  Matthews,  M.J.,  et  al.:  Influence  of  histologic 
subtype  of  small  cell  carcinoma  of  the  lung  on  clincial 
presentation,  response  to  therapy,  and  survival.  JNCI 
65:1225-1230,  1980. 

8.  Hallgrimsson ,  J.:  Lung  tumors  in  Iceland.  Acta.  Path. 

Microbiol.  Scand.  81:813-823,  1973. 

9.  Kyriakos,  M.,  Webber,  B.:  Cancer  of  the  lung  in  young  men. 
Journal  of  Thoracic  and  Cardiovascular  Surgery  67:634-648,  1974. 

10.  Kennedy,  A.:  Lung  cancer  in  young  adults.  Brit.  J.  Dis.  Chest 
66:147-154,  1972. 

11.  Putnam,  J.S.:  Lung  carcinoma  in  young  adults.  JAMA  238:35-36, 
1977. 

12.  Cohen,  M.H.,  Matthews,  M.J.:  Small  cell  bronchogenic 
carcinoma:  A  distinct  clinopathologic  entity.  Seminars  in 
Oncology  5:234-243,  1978. 

13.  Weiss,  W. ,  Boucot,  K.R.,  et  al.:  Risk  of  lung  cancer  according 
to  histologic  subtype  and  cigarette  dosage.  JAMA  222:799-801, 
1972. 


72 


14.  Auerbach,  0.,  Garfinkel,  L.,  Parks,  V.;  Histologic  type  of  lung 
cancer  in  relation  to  smoking  habits,  year  of  diagnosis  and 
sites  of  metastases.  Chest  67:382-387,  1975. 

15.  Johnston-Early,  A.,  Cohen,  M.H.,  et  al.:  Smoking  abstinence  and 
small  cell  cancer  survival.  JAMA  244:2175-2179,  1980. 

16.  Archer,  V.E.,  Saccomanno,  C.,  Jones,  J.H.:  Frequency  of 
diifferent  histologic  types  of  bronchogenic  carcinoma  as  related 
to  radiation  exposure.  Cancer  34:2056-2060,  1974. 

17.  Wurster-Hill ,  D.H.,  Maurer,  L.H.:  Cytogenetic  diagnosis  of 
cancer:  Abnormalities  of  chromosomes  and  polyploid  levels  in 
the  bone  marrow  of  patients  with  small  cell  anaplastic  carcinoma 
of  the  lung.  J.  Natl.  Cancer  Inst.  61:1065-1075,  1978. 

18.  Vindelov,  L.L.,  Hansen,  H.H.,  et  al.:  Clonal  heterogeneity  of 
small-cell  anaplastic  carcinoma  of  the  lung  demonstrated  by 
flow-cytometric  DNA  analysis.  Cancer  Research  40:4295-4300, 
1980. 

19.  Whang-Peng,  J.,  Kao-Shan,  C.S.,  et  al.:  Specific  chromosome 
defect  associated  with  human  small-cell  lung  cancer:  Deletion 
3p  (14-23).  Science  215:181-182,  1982. 

20.  Hattori,  S.,  Matsuda,  M.,  et  al.:  Oat-cell  carcinoma  of  the 
lung.  Clinical  and  morphological  studies  in  relation  to  its 
histogenesis.  Cancer  30:1014-1024,  1972. 

21.  Bensch,  K.G.,  Gordon,  G.B.,  Miller,  L.R.:  Studies  on  the 
bronchial  counterpart  of  the  Kulchitsky  (argentaffin)  cell  and 
innervation  of  bronchial  glands.  J.  Ultrastruct.  Res. 
12:668-686,  1965. 

22.  Greco,  F.A.,  Hainsworth,  J.,  et  al.:  Hormone  production  and 
paraneoplastic  syndromes,  in  Greco  F.A.,  Oldham,  R.K.,  Bunn, 
P.A.,  Jr.  (eds.):  Small  Cell  Lung  Cancer  (Clinical  Oncology 
Monographs).  New  York,  Grune  and  Stratton,  1981,  pp.  177-223. 

23.  Gazdar,  A.F.,  Carney,  D.N.,  et  al.:  Establishment  of 
continuous,  clonable  cultures  of  small-cell  carcinoma  of  the 
lung  which  have  amine  precursor  uptake  and  decarboxylation  cell 
properties.  Cancer  Research  40:3502-3507,  1980. 

24.  Hattori,  S.,  Matsuda,  M.,  et  al . :  Small  cell  carcinoma  of  the 

lung:  Clinical  and  cytomorphological  studies  in  relation  to  its 

response  to  chemotherapy.  Gann  68:321-331,  1977. 


73 


25.  Tischler,  A.S.;  Small  cell  carcinoma  of  the  lung:  cellular 
origin  and  relationship  to  other  neoplasms.  Seminars  in 
Oncology  5:244-252,  1978. 

26.  Frenster,  J.H.,  Herstein,  P.R.:  Gene  de-repression.  N.  Engl. 

J.  Med.  288:1224-1229,  1973. 

27.  Sorenson,  G.D.,  Pettengill,  O.S.,  et  al.:  Hormone  production  by 
cultures  of  small-cell  carcinoma  of  the  lung.  Cancer 
47:1289-1296,  1981. 

28.  Richardson,  R.L.,  Greco,  F.A.,  et  al.:  Tumor  products  and 
potential  markers  in  small  cell  lung  cancer.  Seminars  in 
Oncology  5:253-262,  1978. 

29.  Baylin,  S.B.,  Gazdar,  A.F.:  Endocrine  biochemistry  in  the 
spectrum  of  human  lung  cancer:  Implications  for  the  cellular 
origin  of  small  cell  carcinoma,  in  Greco  F.A.,  Oldham,  R.K., 
Bunn,  P.A.,  Jr.  (eds.):  Small  Cell  Lung  Cancer  (Clinical 
Oncology  Monographs).  New  York,  Grune  and  Stratton,  1981, 

pp.  123-143. 

30.  Carney,  D.N.,  Ihde,  D.C.,  et  al . :  Serum  neuron-specific 

enolase:  A  marker  for  disease  extent  and  response  to  therapy  of 

small-cell  lung  cancer.  Lancet  1:583-585,  1982. 

31.  Bergh,  J.,  Virtanen,  I.,  et  al.:  Classification  of  human 
pulmonary  neoplasms  into  small  cell  and  non-small  cell 
carcinomas  based  on  their  expression  of  intermediate  filaments. 
Proc.  AACR  25:161,  1984. 

32.  Johnson,  T.S.,  Valdivieso,  M. ,  et  al.:  Flow  cytometric  ploidy 
and  proliferative  activity  in  human  small  cell  lung 
carcinomas:  Potential  diagnostic  and  prognostic  features. 

Proc.  AACR  24:124,  1983. 

33.  Fargion,  S.,  Carney,  D.N.,  et  al.:  Antigenic  heterogeneity  in 
small  cell  lung  cancer  (SCLC)  defined  by  15  monoclonal 
antibodies  (MA) .  Proc.  ASCO  2:55,  1983. 

34.  Morstyn,  G.,  Fargion,  S.,  et  al.:  Homogeneity  among  small  cell 
lung  cancer  (SCLC)  cell  lines  (CL)  from  different  metastatic 
sites  (MS)  in  the  same  patient.  Proc.  ASCO  2:197,  1983. 


1 


74 


35.  Mulshine,  J.,  Rosen,  S.,  et  al.:  Immunohistochemical  expression 
of  human  small  cell  lung  cancer  (SCLC)  epitopes  in  malignant  and 
normal  tissue  by  a  panel  of  rodent  monoclonal  antibodies.  Proc. 
AACR  24:221,  1983. 

36.  Doyle,  A.,  Cuttitta,  F.:  Small  cell  lung  cancer  (SCLC)  and 
non-small  cell  lung  cancer  (NSCLC)  lines  differ  markedly  in  the 
expression  of  HLA-frame  and  B-2  microglobulin  (B--M)  antigens. 
Proc.  ASCO  2:12,  1983. 

37.  Doyle,  A.,  Gazdar,  A.,  et  al.:  The  deficit  in  HLA  expression  in 
small  cell  lung  cancer  (SCLC)  resides  at  the  transcriptional 
level  and  is  revised  by  interferon.  Proc.  ASCO  3:52,  1984. 

38.  Carney,  D.N.,  Gazdar,  A.F.,  et  al.:  Two  distinct  classes  of 
tumor  cells  cultured  from  patients  (pts)  with  small  cell  lung 
cancer  (SCLC).  Proc.  ASCO  3:218,  1984. 

39.  Knop,  R.H.,  Carney,  D.N.,  et  al . :  Differentiation  of  small  cell 
lung  cancer  (SCLC)  and  non-small  cell  lung  cancer  (NSCLC)  human 
tumor  cell  lines  (CL)  using  phosphocreatine  (Per)  signals  as 
detected  by  31P  nuclear  magnetic  resonance  (NMR)  spectroscopy. 
Proc.  AACR  25:25,1984. 

40.  Matthews,  M.J.,  Hirsch,  F.R.:  Problems  in  the  diagnosis  of 
small  cell  carcinoma  of  the  lung,  in  Greco,  F.A.,  Oldham,  R.K., 
Bunn,  P.A.,  Jr.  (eds.);  Small  Cell  Lung  Cancer  (Clinical 
Oncology  Monographs).  New  York,  Grune  and  Stratton,  1981, 

pp.  35-50. 

41.  Matthews,  M.J.:  Morphology  of  lung  cancer.  Seminars  in 
Oncology  1:175-182,  1974. 

42.  Hirsch,  F.,  Matthews,  M.,  Yesner,  R.:  Problems  in  the 
histopathologic  classification  of  small  cell  carcinoma  of  the 
lung.  An  interobservational  examination.  Abst .  Int.  Assoc. 
Study  Lung  Cancer,  Copenhagen,  1980,  p.  77. 

43.  Janis,  M,  Matthews,  M.J.,  Bonfiglio,  T.,  et  al.:  Consistency  in 
the  diagnosis  of  small  cell  carcinoma  of  the  lung.  Abst.  Int. 
Assoc.  Study  Lung  Cancer,  Copenhagen,  1980,  p.  79. 

Friesenhahn,  G.M.,  Clark,  G.M.,  et  al.:  Clinical  presentation 
of  689  patients  with  carcinoma  of  the  lung.  Proc.  AACR  24:143, 
1983. 


44. 


75 


45.  Livingston,  R.B.,  Trauth,  C.J.,  Greenstreet,  R.L.:  Small  cell 
carcinoma:  Clinical  manifestations  and  behavior  with  treatment, 
in  Greco,  F.A.,  Oldham,  R.K.,  Bunn,  P.A.,  Jr.  (eds.):  Small 
Cell  Lung  Cancer  (Clinical  Oncology  Monographs).  New  York,  Grune 
and  Stratton,  1981,  pp.  285-300. 

46.  Ihde,  D.C.,  Makuch,  R.W.,  et  al.;  Prognostic  implications  of 
stage  of  disease  and  sites  of  metastases  in  patients  with  small 
cell  carcinoma  of  the  lung  treated  with  intensive  combination 
chemotherapy.  Am.  Rev.  Respir.  Dis.  123:500-507,  1981. 

47.  Zelen,  M.:  Keynote  address  on  biostatistics  and  data 
retrieval.  Cancer  Chemotherapy  Reports,  Part  3,  4:31-42,  1973. 

48.  Ihde,  D.C.,  Hansen,  H.H.:  Staging  procedures  and  prognostic 
factors  in  small  cell  carcinoma  of  the  lung  in,  Greco,  F.A., 
Oldham,  R.K.,  Bunn,  P.A.,  Jr.  (eds.):  Small  Cell  Lung  Cancer 
(Clinical  Oncology  Monographs).  New  York,  Grune  and  Stratton, 

1981,  pp.  261-283. 

49.  Livingston,  R.B.,  Moore,  T.N.,  et  al.:  Small-cell  carcinoma  of 
the  lung:  combined  chemotherapy  and  radiation.  Annals  of 
Internal  Medicine  88:194-199,  1978. 

50.  Chang,  A.E.,  Schaner,  E.G.,  et  al . :  Evaluation  of  computed 
tomography  in  the  detection  of  pulmonary  metastases.  Cancer 
43:913-916,  1979. 

51.  Underwood,  G.H.,  Hooper,  R.G.,  et  al.:  Computed  tomographic 
scanning  of  the  thorax  in  the  staging  of  bronchogenic 
carcinoma.  N.  Engl.  J.  Med.  300:777-778,  1979. 

52.  McCrea,  E.S.,  Fuks,  J.Z.,  et  al.:  Computed  tomography  of  the 
chest  in  staging  and  follow-up  of  small  cell  lung  cancer  (SCLC). 
Proc.  ASCO  2:195,  1983. 

53.  Harper,  P.G.,  Souhami,  R.L.,  et  al.:  Tumor  size,  response  rate, 
and  prognosis  in  small  cell  carcinoma  of  the  bronchus  treated  by 
combination  chemotherapy.  Cancer  Treatment  Reports  66:463-470, 

1982. 

54.  Shepherd,  F.A.,  Ginsberg,  R.,  et  al . :  "Very  limited"  small  cell 
lung  cancer  (SCLC) — results  of  non-surgical  treatment.  Proc. 
ASCO  3:223,  1984. 

55.  Compling,  B.,  Quirt,  I.,  et  al.:  Bone  marrow  involvement  in 
small  cell  lung  cancer  (SCLC).  Proc.  ASCO  2:195,  1983. 


t 


76 


56.  Byrne,  R.,  Levitan,  N.,  et  al.:  The  value  of  the  radionuclide 
bone  scan  and  bone  marrow  biopsy  in  staging  small  cell  lung 
cancer  (SCLC).  Proc.  ASCO  2:189,  1983. 

57.  Osterlind,  K.,  Ihde,  D.C.,  et  al.:  Staging  and  prognostic 
factors  in  small  cell  carcinoma  of  the  lung.  Cancer  Treatment 
Reports  67:3-9,  1983. 

58.  Kramer,  B.S.,  Wozniak,  A.J.,  Ross,  W.E.:  A  retrospective  study 
of  the  value  of  staging  procedures  in  patients  (Pts)  with  small 
cell  undifferentiated  cancer  of  lung  (SCCL) .  Proc.  ASCO  1:2, 

1982. 

59.  Feld,  R.,  Deboer,  G.,  Evans,  W.K.:  Prognostic  factors  in 
patients  with  small  cell  carcinoma  of  the  lung  (SCCL).  Proc. 

AACR  2A:145,  1983. 

60.  Hirsch,  F.R.,  Hansen,  H.H.:  Bone  marrow  involvement  in  small 
cell  anaplastic  carcinoma  of  the  lung.  Cancer  46:206-211,  1980. 

61.  Mulshine,  J.L.,  Makuch,  R.W.,  et  al . :  Diagnosis  and 
significance  of  liver  metastases  in  small  cell  carcinoma  of  the 
lung.  Journal  of  Clinical  Oncology  2:733-741,  1984. 

62.  Mulshine,  J.,  Matthews,  J.,  et  al.:  Staging  evaluation  of  the 
liver  in  small  cell  cancer  of  the  lung  (SCCL).  Proc.  ASCO  1:140, 

1982. 

63.  Hansen,  S.W.,  Jensen,  F.,  et  al . :  Staging  procedures  of  the 
liver  in  small  cell  bronchogenic  carcinoma  (SCC).  A  prospective 
comparative  trial  of  peritoneoscopy  with  liver  biopsy  versus 
ultrasonography  with  fine  needle  aspiration.  Proc.  ASCO  2:5, 

1983. 

64.  Levitan,  N.,  Byrne,  R.,  et  al . :  Computerized  cranial  tomography 
(CCT)  in  small  cell  lung  cancer.  Proc.  ASCO  2:189,  1983. 

65.  Aisner,  J.,  Alberto,  P.,  et  al.:  Role  of  chemotherapy  in  small 
cell  lung  cancer:  A  consensus  report  of  the  international 
association  for  the  study  of  lung  cancer  workshop.  Cancer 
Treatment  Reports  67:37-43,  1983. 

66.  Dewys,  W.D.,  Begg,  C.,  et  al . :  Prognostic  effect  of  weight  loss 
prior  to  chemotherapy  in  cancer  patients.  American  Journal  of 
Medicine  69:491-497,  1980. 


77 


67.  Poplin,  E.,  Whitacre,  M.,  et  al.;  Small  cell  carcinoma  of  the 
lung  (SCCL):  Influence  of  age  on  treatment  outcome.  Proc.  ASCO 
3:223,  1984. 

68.  Sculier ,  J.P.,  Feld,  R.,  et  al.:  CEA:  A  useful  prognostic 
marker  for  small  cell  lung  cancer  (SCLC).  Proc.  AACR  25:158, 
1984. 

69.  Koretz,  M.M.,  Carter,  S.K.:  Laboratory  parameters  as 
alternative  to  performance  status  in  predicting  survival  in  oat 
cell  carcinoma.  Proc.  ASCO  1:154,  1982. 

70.  Cohen,  M.H.,  Makuch,  R.,  et  al . :  Laboratory  parameters  as  an 
alternative  to  performance  status  in  prognostic  stratification 
of  patients  with  small  cell  lung  cancer.  Cancer  Treatment 
Reports  65:187-195,  1981. 

71.  Davis,  S.,  Stanley,  K.E.,  et  al.:  Small  cell  carcinoma  of  the 

lung:  Survival  according  to  histologic  subtype.  Cancer 

47:1863-1866,  1981. 

72.  Carney,  D.N.,  Matthews,  M.J.,  et  al . :  Influence  of  histologic 
subtype  of  small  cell  carcinoma  of  the  lung  on  clinical 
presentation,  response  to  therapy,  and  survival.  J.  Natl. 

Cancer  Inst.  65:1225-1230,  1980. 

73a.  DeFusco,  P.,  Cole,  S.,  et  al . :  The  effect  of  survival  of 

histopathologic  subtypes  of  small  cell  lung  cancer  (SCLC):  A 
community  hospital  experience.  Proc.  ASCO  3:216,  1984. 

73b.  Yesner,  R.  Personal  communication. 

73c,  Byhardt,  R.W.,  Cox,  J.D.:  Is  chest  radiotherapy  necessary  in 

any  or  all  patients  with  small  cell  carcinoma  of  the  lung?  Yes. 
Cancer  Treatment  Reports  67:209-221,  1983. 

74.  Selawry,  O.S.:  Monochemotherapy  of  bronchogenic  carcinoma  with 
special  reference  to  cell  type.  Cancer  Chemotherapy  Reports, 
Part  3,  4:177-188,  1973. 

75.  Shackney,  S.E.,  Straus,  M.J.,  Bunn,  P.A.,  Jr.:  The  growth 
characteristics  of  small  cell  carcinoma  of  the  lung,  in  Greco, 
F.A.,  Oldham,  R.K.,  Bunn,  P.A.,  Jr.  (eds.):  Small  Cell  Lung 
Cancer  (Clinical  Oncology  Monographs).  New  York,  Grune  and 
Stratton,  1981,  pp.  225-234. 


78 


76.  Cox,  J.D.,  Byhardt,  R.W.,  et  al.:  Dose-time  relationships  and 
the  local  control  of  small  cell  carcinoma  of  the  lung. 

Radiology  128:205-207,  1978. 

77.  Ginsberg,  S.J.,  Comis,  R.L.,  et  al.:  Long-term  survivorship  in 
small-cell  anaplastic  lung  carcinoma.  Cancer  Treatment  Reports 
63:1347-1349,  1979. 

78.  Muggia,  F.M.,  Krezoski,  S.K.,  Hansen,  H.H.:  Cell  kinetic 
studies  in  patients  with  small  cell  carcinoma  of  the  lung. 
Cancer  34:1683-1690,  1974. 

79.  Comis,  R.L.:  Small  cell  carcinoma  of  the  lung.  Cancer 
Treatment  Reviews  9:237-258,  1982. 

80.  Brigham,  B.A.,  Bunn,  P.A.,  Jr.,  et  al.:  Growth  rates  of  small 
cell  bronchogenic  carcinomas.  Cancer  42:2880-2886,  1978. 

81.  Abeloff,  M.D.,  Eggleston,  J.C.,  et  al.:  Changes  in  morphologic 
and  biochemical  characteristics  of  small  cell  carcinoma  of  the 
lung.  American  Journal  of  Medicine  66:757-764,  1979. 

82.  Matthews,  M.J.,  Ihde,  D.C.,  et  al . :  Effects  of  chemotherapy  on 
the  histology  of  small  cell  carcinoma  of  the  lung  (SCCL) — a 
postmortem  study  of  54  cases.  Proc .  AACR-ASCO  20:397,  1978. 

83.  Matthews,  M.J.,  Rozencweig,  M.,  et  al.:  Long-term  survivors 
with  small  cell  carcinoma  of  the  lung.  Europ.  J.  Cancer 
16:527-531,  1980. 

84.  Johnson,  B.E.,  Ihde,  D.C.,  et  al.:  Five  to  ten  year  follow-up 
of  small  cell  lung  cancer  (SCLC)  patients  disease  free  at  30 
months:  Chronic  toxicities  and  late  relapses.  Proc.  ASCO 
3:218,  1984. 

85.  Kalter,  S.,  Farha,  P.,  et  al.:  Long  term  survivors  with 
small-cell  lung  cancer  (SCLC):  The  M.D.  Anderson  (MDA) 
experience  from  1972-1980.  Proc.  ASCO  3:229,  1984. 

86.  Livingston,  R.B.:  Long-term  survivors  with  small-cell  lung 
cancer  (SCLC):  A  southwest  oncology  group  (SWOG)  study.  Proc. 
ASCO  2:187,  1983. 


79 


87.  Greco,  F.A.,  Einhorn,  L.H.,  et  al.:  Small  cell  lung 
cancer:  Progress  and  perspectives.  Seminars  in  Oncology 
5:323-335,  1978. 

88.  Roswit,  B.,  Patno ,  M.E.,  et  al.:  The  survival  of  patients  with 
inoperable  lung  cancer:  A  large-scale  randomized  study  of 
radiation  therapy  versus  placebo.  Radiology  90:688-697,  1968. 

89.  Matthews,  M.J.,  Kanhouwa,  S.,  et  al.:  Frequency  of  residual  and 
metastatic  tumor  in  patients  undergoing  curative  surgical 
resection  for  lung  cancer.  Cancer  Chemotherapy  Reports,  Part  3, 
4:63-67,  1973. 

90.  Fox,  W.,  Scadding,  J.G.:  Medical  research  council  comparative 
trial  of  surgery  and  radiotherapy  for  primary  treatment  of 
small-celled  or  oat-celled  carcinoma  of  bronchus.  Ten-year 
follow-up.  Lancet  2:63-65,  1973. 

91 .  Medical  Research  Council  Lung  Cancer  Working 

Party:  Radiotherapy  alone  or  with  chemotherapy  in  the  treatment 

of  small-cell  carcinoma  of  the  lung.  Br.  J.  Cancer  40:1-10, 
1979. 

92.  Greco,  F.A.,  Oldham,  R.K.:  Clinical  management  of  patients  with 
small  cell  lung  cancer  in,  Greco,  F.A.,  Oldham,  R.K.,  Bunn, 

P.A.,  Jr.  (eds.):  Small  Cell  Lung  Cancer  (Clinical  Oncology 
Monographs).  New  York,  Grune  and  Stratton,  1981,  pp.  353-379. 

93.  Bunn,  P.A.,  Cohen,  M.H. ,  et  al.:  Advances  in  small  cell 
bronchogenic  carcinoma.  Cancer  Treatment  Reports  61:333-342, 
1977. 

94.  Livingston,  R.B.,  Moore,  T.N.,  et  al.:  Small  cell  carcinoma  of 
the  lung:  Combined  chemotherapy  and  radiation:  A  Southwest 
Oncology  Group  Study.  Ann.  Intern.  Med.  88:194-199,  1978. 

95.  Papac,  R.,  Bien  R.,  et  al.:  Combined  modality  treatment  of 
small  cell  lung  cancer  (SCLC)  with  cyclophosphamide  (CYT), 

VP-16,  methotrexate  (MTX)  and  high  dose  radiation.  Proc.  ASCO 
3:216,  1984. 

96.  Goodman,  G.E.,  Miller,  T.P.,  et  al.:  Treatment  of  small  cell 
lung  cancer  with  VP-16,  vincristine,  doxorubicin  ( Adriamycin) , 
cyclophosphamide  (EVAC),  and  high-dose  chest  radiotherapy. 
Journal  of  Clinical  Oncology  1:483-488,  1983. 


80 


97.  Zekan,  P.,  Jackson,  D.,  et  al.:  Cyclophosphamide,  Adriamycin 
and  vincristine  (CAV)  versus  VP-16-213  +  CAV  (VCAV)  in  the 
treatment  of  small  cell  carcinoma  of  the  lung  (SCC).  Proc.  ASCO 
2:193,  1983. 

98.  Livingston,  R.B.,  Mira,  J.G.,  et  al . ;  Combined  modality 
treatment  of  extensive  small  cell  lung  cancer:  A  Southwest 
Oncology  Group  study.  Journal  of  Clinical  Oncology  2:585-590, 
1984. 

99.  Baglan,  R.J.,  Marks,  J.E.:  Comparison  of  symptomatic  and 
prophylactic  irradiation  of  brain  metastases  from  oat  cell 
carcinoma  of  the  lung.  Cancer  47:41-45,  1981. 

100.  Rosen,  S.T.,  Makuch,  R.W.,  et  al.:  Role  of  prophylactic  cranial 
irradiation  in  prevention  of  central  nervous  system  metastases 
in  small  cell  lung  cancer.  Potential  benefit  restricted  to 
patients  with  complete  response.  American  Journal  of  Medicine 
74:615-624,  1983. 

101.  Cox,  J.D.,  Komaki,  R.,  et  al.:  Results  of  whole-brain 
irradiation  for  metastases  from  small  cell  carcinoma  of  the 
lung.  Cancer  Treat.  Rep.  64:957-961,  1980. 

102.  Crane,  J.,  Lichter,  A,  et  al.:  Therapeutic  cranial  radiotherapy 
(RT)  for  brain  metastases  in  small  cell  lung  cancer  (SCLC). 

Proc.  AACR  24:145,  1983. 

103.  Sculler,  J.P.,  Feld,  R.,  et  al.:  Neurological  complications  in 
patients  (Pts)  with  small  cell  lung  cancer  (SCLC).  Proc.  ASCO 
3:222,  1984. 

104.  Ellison,  N.,  Bernath,  A.,  et  al . :  Disturbing  problems  of 
success:  Clinical  status  of  long-term  survivors  of  small  cell 
lung  cancer  (SCLC).  Proc.  ASCO  1:149,  1982. 

105.  Scher,  H.,  Hilaris,  B.,  Wittes,  R.:  Long  term  follow-up  of 
combined  modality  therapy  in  small  cell  carcinoma  of  the  lung. 
Proc.  ASCO  2:199,  1983. 

106.  Looper,  J.D.,  Einhorn,  L.H.,  et  al.:  Severe  neurologic  problems 
following  successful  therapy  for  small  cell  lung  cancer  (SCLC). 
Proc.  ASCO  3:231,  1984. 


81 


107.  Craig,  J.,  Jackson,  D.,  et  al . :  Prospective  evaluation  of 
changes  in  computerized  cranial  tomography  (CCT)  in  patients 
with  small  cell  carcinoma  (SCLC)  treated  vi^ith  chemotherapy  and 
cranial  irradiation.  Proc.  ASCO  3:224,  1984. 

108.  Catane,  R.,  Lichter,  A.,  et  al.:  Small  cell  lung 

cancer:  Analysis  of  treatment  factors  contributing  to  prolonged 
survival.  Cancer  48:1936-1943,  1981. 

109.  Cox,  J.D.,  Byhardt,  R.W.,  et  al . :  Dose-time  relationships  and 
the  local  control  of  small  cell  carcinoma  of  the  lung. 

Radiology  128:205-207,  1978. 

110.  Son,  Y.H.,  Kapp,  D.S.:  Esophago-pulmonary  toxicity  from 
concomitant  use  of  Adriamycin  and  irradiation.  Connecticut 
Medicine  45:755-759,  1983. 

111.  Bleehen,  N.M.,  Bunn,  P.A.,  et  al.:  Role  of  radiation  therapy  in 
small  cell  anaplastic  carcinoma  of  the  lung.  Cancer  Treatment 
Reports  67:11-19,  1983. 

112.  Byhardt,  R.W.,  and  Cox,  J.D.:  Is  chest  radiotherapy  necessary 
in  any  or  all  patients  with  small  cell  carcinoma  of  the  lung? 
Yes.  Cancer  Treatment  Reports  67:209-215,  1983. 

113.  Cohen,  M.H.:  Is  thoracic  radiation  necessary  for  patients  with 
limited-stage  small  cell  lung  cancer?  No.  Cancer  Treatment 
Reports  67:217-221,  1983. 

114.  Comis,  R.L.:  Small  cell  carcinoma  of  the  lung.  Cancer 
Treatment  Reviews  9:237-258,  1982. 

115.  Peschel ,  R.E.,  Kapp,  D.S.,  et  al.:  Long  term  survivors  with 
small  cell  carcinoma  of  the  lung.  Int.  J.  Radiation  Biol.  Phys. 
7:1545-1548,  1981. 

116.  Hansen,  H.H.,  Dombernowsky ,  P.,  et  al . :  Chemotherapy  versus 
chemotherapy  plus  radiotherapy  in  regional  small-cell  carcinoma 
of  the  lung — a  randomized  trial.  Proc.  AACR  20:277,  1979. 

117.  Bunn,  P.,  Cohen,  M. ,  et  al.:  Randomized  trial  of  chemotherapy 
versus  chemotherapy  plus  radiotherapy  in  limited  stage  small 
cell  lung  cancer  (SCLC).  Proc.  ASCO  2:200,  1983. 


82 


118.  Perez,  C.A.,  Einhorn,  L.,  et  al . :  Preliminary  report  on  a 
randomized  trial  of  radiotherapy  (RT)  to  the  thorax  in  limited 
small  cell  carcinoma  of  the  lung  treated  with  multiagent 
chemotherapy.  Proc.  ASCO  2:190,  1983. 

119.  Mira,  J.G.,  Kies,  M.S.,  et  al.:  Influence  of  chest  radiotherapy 
(RT)  in  response,  remission  duration,  and  survival  in 
chemotherapy  (CT)  responders  in  localized  small  cell  lung 
carcinoma  (SCLC):  A  Southwest  Oncology  Group  (SWOG)  study. 

Proc.  ASCO  3:212,  1984. 

120.  Abeloff ,  M.D.,  Klastersky,  J.,  et  al.:  Complications  of 
treatment  of  small  cell  carcinoma  of  the  lung.  Cancer  Treatment 
Reports  67:21-26,  1983. 

121a.  Aisner ,  J.,  Wiernik,  P.H.:  Complications  of  treatment  and  of 
improved  survival  in  patients  with  small  cell  carcinoma  of  the 
lung,  in  Greco,  F.A.,  Oldham,  R.K.,  Bunn,  P.A.,  Jr.:  Small  Cell 
Lung  Cancer  (Clinical  Oncology  Monographs).  New  York,  Grune  and 
Stratton,  1981,  pp.  381-398. 

121b.  Rubin,  P.H.,  Scarantino,  C.H.:  The  bone  marrow  organ:  The 
critical  structure  in  radiation/drug  interaction.  Int.  J. 
Radiat.  Oncol.  Biol.  Phys.  4:3-23,  1978. 

122.  Comis,  R.,  Meyer,  J.,  et  al . :  The  current  results  of 
chemotherapy  (CTH)  plus  adjuvant  surgery  (AS)  in  limited  small 
cell  anaplastic  lung  cancer  (SCALC).  Proc.  ASCO  1:147,  1982. 

123.  Comis,  R.,  Meyer,  J.,  et  al . :  The  impact  of  TNM  stage  on 
results  with  chemotherapy  (CT)  and  adjuvant  surgery  (AS)  in 
small  cell  lung  cancer  (SCLC).  Proc.  ASCO  3:226,  1984. 

124.  Foster,  J.M.,  Prager,  R.L.,  et  al.:  Limited-stage  small-cell 
carcinoma  (SCC):  Prospective  evaluation  regarding  the 
feasibility  of  adjuvant  surgery.  Proc.  ASCO  2:196,  1983. 

125.  Friess,  G.,  McCracken,  J.,  et  al . :  Improved  long  term  survival 
associated  with  surgery  in  limited  small  cell  cancer  of  the  lung 
(SCCL).  A  Southwest  Oncology  Group  (SWOG)  study.  Proc.  ASCO 
3:219,  1984. 

126.  Ihde,  D.C.,  Lichter,  A.S.,  et  al.:  Late  intensive  combined 
modalitiy  therapy  (LIRX)  with  autologous  bone  marrow  (ABM) 
infusion  in  extensive  stage  small  cell  lung  cancer  (SCLC).  Proc. 
ASCO  2:198,  1983. 


83 


127.  Valdivieso,  M. ,  Cabanillas,  F.,  et  al.:  Effects  of  intensive 
induction  chemotherapy  for  extensive-disease  small  cell 
bronchogenic  carcinoma  in  protected  environment-prophylactic 
antibiotic  units.  American  Journal  of  Medicine  76:405-412, 

1984. 

128.  Smith,  I.E.,  Evans,  B.D.,  Har^and,  S.J.:  High  dose 
cyclophosphamide  (HDC)  (7  G/M  )  +  autologous  bone  marrow  rescue 
(ABMR)  ofter  conventional  chemotherapy  in  patients  with  small 
cell  lung  cancer  (SCLC).  Proc.  ASCO  2:186,  1983. 

129.  Aroney,  R.S.,  Bell,  D.R.,  et  al . :  Alternating 
non-cross-resistant  combination  chemotherapy  for  small  cell 
anaplastic  carcinoma  of  the  lung.  Cancer  49:2449-2454,  1982. 

130.  Evans,  W.K.,  Feld,  R.,  et  al.:  VP-16  alone  and  in  combination 
with  cisplatin  in  previously  treated  patients  with  small  cell 
lung  cancer.  Cancer  53:1461-1466,  1984. 

131.  Cohen,  M.H.,  Ihde,  D.C.,  et  al.:  Cyclic  alternating  combination 
chemotherapy  for  small  cell  bronchogenic  carcinoma.  Cancer 
Treatment  Reports  63:163-170,  1979. 

132.  Johnson,  D.H.,  Wolff,  S.N.,  et  al . :  High-dose  VP-16-213  (HDE) 
treatment  of  extensive  small  cell  lung  cancer  (SCC).  Proc.  ASCO 
2:193,  1983. 

133.  Woods,  R.L.,  Fox,  R.M.,  Tattersall,  M.H.N.:  Treatment  of  small 
cell  bronchogenic  carcinoma  with  VM-26.  Cancer  Treatment  Reports 
63:2011-2013,  1979. 

134.  Natale,  R.B.,  Gralla,  R.J.,  Wittes,  R.E.:  Phase  II  trial  of 
vindesine  in  patients  with  small  cell  lung  carcinoma.  Cancer 
Treatment  Reports  65:129-131,  1981. 

135.  Petruska,  P.,  Luedke,  D.,  et  al . :  Phase  II  study  of  vindesine 
(v)  in  refractory  small  cell  carcinoma  of  the  lung  (SCC).  Proc. 
AACR/ASCO  21:500,  1981. 

136.  Gregor,  A.,  Cornbleet,  M. ,  et  al.:  Vindesine  and  VP-16-213  as 
primary  therapy  for  poor  prognosis  small  cell  carcinoma  of 
bronchus  (SCCB).  Proc.  ASCO  2:186,  1983. 

137.  Schabel,  F.M.,  Trader,  M.W.,  et 

al.:  Cis-dichlorodiammineplatinum  (11):  Combination 

chemotherapy  and  cross-resistance  studies  with  tumors  of  mice. 
Cancer  Treatment  Reports  63:1459-1473,  1979. 


84 


138.  Livingston,  R.,  Mira,  J.:  Extensive  small  cell  lung 

cancer:  Combined  alkylators  for  induction  with  cisplatin  + 

VP-16  consolidation.  A  Southwest  Oncology  Group  study.  Proc. 
ASCO  3:210,  1984. 

139.  Zekan,  P.,  Jackson,  D.,  et  al . :  Cyclophosphamide,  Adriamycin, 
and  Vincristine  (CAV)  versus  VP-16-213  +  CAV  (VCAV)  in  the 
treatment  of  small  cell  carcinoma  of  the  lung  (SCC).  Proc.  ASCO 
2:193,  1983. 

140.  Messeih,  A.,  Schweitzer,  J.M.,  et  al . , :  The  addition  of 
VP-16-213  to  Cytoxan,  Adriamycin  and  Vincristine  for  remission 
induction,  and  survival  in  patients  with  small  cell  lung 
cancer.  Proc.  ASCO  2:201,  1983. 

141.  Rome,  L.S.,  Portlock,  C.S.,  et  al . :  Cyclophosphamide, 
Adriamycin,  vincristine  (CAV)  vs.  cyclophosphamide,  Adriamycin, 
vincristine  alternating  with  VP-16  (Etoposide)  (CAV/E)  in  the 
treatment  of  small  cell  cancer  (SCC)  of  the  lung.  Proc.  ASCO  4, 
1985  (in  press). 


Ex-cigarette 

smokers 


<1  pack 
a  day 


1-2  packs 
a  day 


14.5% 


19.2% 


23.9% 


Total  subjects  =  163 


From; 


Auerbach,  0., 


Garf inkel ,  L. , 


and  Parks, 


V.R. 


[14; 


Table  _1  —  Age  Standardized  Percentage  Distribution  of 
Cigarette  Smoking  Habit 


2+  packs 
a  day 


31.1% 


SCCL  by 


vr 


Exposure  Group+ 


Excess  Cases 


1  -  359 

8.27* 

360  -  1779 

22.07* 

>  1800 

33.69* 

Combined  groups 

64.03* 

=  significantly  different  from  expected  number  of  cases  (£.<.01) 


+Exposure  is  quantified  by  "Working  Level  Month"  (WLM)  Groups.  One 
WLM  is  a  month's  work  performed  in  an  atmosphere  containing  a  standard 
radiation  dose  per  liter  of  air. 


From: 


Archer,  J.E.,  Saccomano,  G., 


and  Jones, 


J.H. 


[16] 


Table  2.  —  Distribution  of  Excess  (Presumably  Radiation-Induced) 
Bronchogenic  Cancers  by  Radiation  Exposure  Group 


87 


Symptom 


Percentage  of  patients  with  the  symptom 


Cohen  and  Matthews 


[12] 


Friesenhahn , 


et  al. 


[44] 


Cough 

76 

37 

Chest  pain 

36 

28 

Dyspnea 

34 

31 

Pneumonitis 

25 

NR 

Wheeze 

22 

NR 

Hemoptysis 

15 

17 

Fatigue 

NR 

21 

Hoarseness 

15 

NR 

SVC  syndrome 

12 

NR 

NR  =  Not  Reported 


Table  _3  —  Symptoms  of  SCCL 


88 


At  Presentation^ At  Autopsy^ 

( total  pts  =  375)  ( total  pts  =  163) 


Liver 

32 

61.7 

Bone 

30 

35 

Bone  marrow 

16 

NR 

Brain 

14 

50 

Skin,  soft  tissue, 
nodes 

16 

75.5  (excluding 

"chest  wall") 

Effusion/pleura 

15 

22.7 

Heart 

NR 

20.3 

NR  =  Not  Reported 


[45  ] 

From:  Livingston,  R.B.,  Trauth,  C.J.,  Greenstrget,  R.L.  and 
Auerbach,  0.,  Garfinkel,  L. ,  Parks,  V.  ^ 


Table  _4  —  Percent  Distribution  of  Metastases  at  Presentation  and 
at  Autopsy  in  Two  Studies 


From 


d1 


Site 


Procedure 


Refommended 


Primary 


Mediant  inum 


Bone  marrow 


Liver 


Chest  X-ray 
Fiberoptic  bronchoscopy 

Mediastinoscopy* 
Gallium  scan 

Biopsy  and  aspiration 
Bilateral  biopsies 
Scintifrrams 

Peritoneoscopy  and 
liver  biopsy 
Ultra  so  no^aphy 
CT  scan 


Lymph  nodes  and  skin  Fine-needle  aspiration 


CSS 


Retropentoneal 

organs 


CT  scans 
Scintigrams 
Lumbar  puncture 
Myelograms 

CT  scans 

Ultrasonography 

Laparotomy 


4 


4 


4 


If  poiiitive 
initially 


If  signs' 
symptoms 


4 


•Whenever  possible. 


Osterlind  K.,  Ihde,  D.C.  et  al 


[57] 


Table  _5  —  Recommendations  for  Restaging 


Definite 

Stage  of  disease 

Performance  status 

Probable 

Liver  or  CNS  metastases 

Laboratory  parameters 

Possible 

Weight  loss 

Number  of  metastatic  sites 

Age 

Sex 

Size  of  lesion  ("very  limited"  vs.  other) 

None 

Histologic  subtype  (1977  WHO  classification) 

Invest isational 

Histologic  subtype  (small  cell-large  cell  vs 
classic  small  cell)+ 

Adapted  from  Ihde  D.C., 

and  Hansen,  H.H.[48] 

+Proposed  by  pathology  panel  of  the  International  Association  for  the 
Study  of  Lung  Cancer. [73b] 


Table  ^  —  Prognostic  Factors  in  SCCL 


91 


Number 

Complete 

Median 

Survival 

of 

Pts 

Response 

(weeks) 

Treatment 

LD 

ED 

LD 

ED 

LD 

ED 

Placebo^^^^ 

38 

108 

+ 

+ 

11.7 

5.0 

Placebo 

29 

+ 

+ 

+ 

>16 

+ 

Surgery^^®^ 

68 

+ 

+ 

+ 

28.5^ 

+ 

Radiation^ 

70 

+ 

*«• 

+ 

44^ 

+ 

D  [88] 

Radiation 

53 

+ 

+ 

>16 

+ 

CAV  +  RT^^^^ 

108 

250 

41% 

14% 

52 

26 

CME  +  RT^^^^ 

(38  LD 

+  ED) 

86% 

27% 

56 

20 

CAVE  +  RT^^^^ 

33 

11 

76% 

34% 

92 

36 

CAVE  +  RT^^"^^ 

28 

29 

61% 

21% 

60 

37 

MEV/  CAV/^^^^ 

MEV  -  CAV 

+ 

453 

+ 

16% 

+ 

31 

+  =  Inapplicable 

C  = 

cyclophos 

phamide 

M  =  methotrexate 

=  Not  Reported 

A  = 

Adriamycin 

RT  =  Radiation 

Therapy 

a  =  Mean  Survival 

V  = 

vincristine 

E  =  Etoposide 

(VP  16-213) 


Table  7  —  Treatment  Results  in  SCCL:  Selected  Studies 


92 


Number  of 


Treatment 

Patients 

o  [90] 

Surgery 

58 

Radiation^ 

70 

Chemotherapy  ±  RT^^^^ 

255 

CAV  +  RT^^^^ 

400 

Patient  Long-Term 

Characteristics  Survivors 


All 

LD 

0% 

LD 

All 

LD 

5% 

LD 

+ 

6% 

LD  + 

ED 

100 

LD 

4% 

(LD  + 

ED) 

300 

ED 

11% 

LD 

2% 

ED 

+  =  Not  Reported  RT  =  Radiation  Therapy 

C  =  cyclophosphamide  A  =  Adriamycin  (doxorubicin) 

V  =  vincristine 


Table  8  —  Long-Term  Survival  (>_5  years)  in  SCCL 


Treitntfit  Sctifwi 
GROUP  I  -  LfmfM  Disease 


I  oe  Lu  i/>  t—  <  o 


«  u  > 


«<(■>> 

«e  <  u»> 


tf)  «cu  > 


•CU>pr> 


I 


%.  MCC 
•—  *5  H-  <-> 
^  U  > 


T»<« 

3>-  u  ^ 

o  > 


Ta b  1  e  9  —  Treatirient  Protocol  for  Limited  Disease 


1 


I 


I 


Treatment  Schema 


GROUP  2  •  Extensive  Disease 


cycle 


1 

2 

3 

4 

5 

6  7 

8 

9 

Adri  a 

A 

A 

A 

A 

A  A 

A 

A 

Diagnosis 

CTX 

C 

C 

C 

C 

C  C 

C 

C 

- >^VCR 

V 

V 

V 

V 

V  V 

V 

V 

Elective 

Rad  Rx 

Prophylactic 

j 

b 

Whole  Brain 

Rad 

CTX 

VCR 


Adri  a  A  A  A  A 

CTX  VP-16  C  VP-16  C  VP-16  C  VP-16  C  VP-16 

VCR  V  V  V  V 

1  2,  3  H  S 

cycle 


R 

E 

S 

T 

A 

G 

E 


2 

Adri a  :  Adriamycin  40  mg/m  IV  day  1 

2 

CTX  :  Cyclophosphamide  1000  mg/m  IV  day  1 

2 

VCR  :  Vincristine  1.4  mg/m  IV  day  1  (each  dose  limited  to  2  mg  total) 
VP-16  125  mg/m^  IV  days  1,  3,  5 


Table  10  —  Treatment  Protocol  for  Extensive  Disease 


CAV  Therapy  Patients 


n  =  21  (54%) 


10  females:  11  males 
Median  age  (range):  59  years  (49-71) 
Subgroup :  CAV  -  Limited  Disease  Patients 


n  =  8  (38%) 


P.S.  0  =  1 - - 

— - ^6  pts.  fully  am 

P.S.  1  =  5 - 

^ (75%) 

P.S.  2  =  0  - _ _ 

1  pt.  not  fully 

P.S.  3  =  1 

am  b  u  1  a  t  o  r  y 

P.S.  4  =  0^- — 

(12%) 

P.S.  unknown  =  1 

Subgroup :  CAV  -  Extensive  Disease  Patients 


n  =  13  (62%) 


P.S.  0  =  0 - 

■—-——^8  pts.  fully  ambulatory 

P.S.  1  =  8 - ' 

(62%) 

P.S.  2  =  4^^ 

5  pts.  not  fully 

P.S.  3=0 

ambulatory 

P.S.  4  =  1^^ 

(38%) 

CAV/E  Therapy  Patients  n  =  18  (46%) 

10  females:  8  males 

Median  age  (range):  66  years  (53-72) 

Subgroup :  CAV/E  -  Limited  Disease  Patients  n  =  7  (39%) 


P.S.  0=3 
P.S.  1  =  2 

P.S.  2=2 
P.S.  3=0 
P.S.  4=0 


5  pts.  fully  ambulatory 
(71%) 

2  pts.  not  fully 

ambulatory 

(29%) 


Subgroup :  CAV/E  -  Extensive  Disease  Patients  n  =  11  (61%) 


P.S.  0  =  - - 

- — -—^3  pts.  fully  ambulatory 

P.S.  1  =  2^-— 

■ - ^  (27%) 

P.S.  2  =  4-^^ _ 

8  pts.  not  fully 

P.S.  3=3 

ambulatory 

P.S.  4  =  1— ^  ■ 

(73%) 

E  =  Etoposide  (VP-16-213) 

A  =  Adriamycin  (doxorubicin) 


C  =  cyclophosphamide 
V  =  vincristine 


Table  11  —  Patient  Characteristics  by  Treatment  Arm 


96 


Number  of  pts 

%  of 

ED  pts 

with  disease 

with 

disease 

Number  of  pts  with 

at 

site+ 

at  site 

this  site  as  only 
involvement  beyond 

Site 

CAV 

CAV/E 

CAV 

CAV/E 

primary  tumor 

Liver 

8 

6 

62% 

55% 

3 

Bone 

7 

5 

54% 

45% 

1 

Bone  Marrow 

6 

1 

46% 

9% 

2 

Nodes  (excluding  chest) 

3 

2 

23% 

18% 

- 

Pleura 

2^:- 

2.VX- 

15% 

18% 

- 

Bilateral  Lung 

1 

1 

8% 

9% 

- 

Subcutaneous  Nodules 

2 

- 

.5% 

- 

- 

Brain 

- 

1 

- 

9% 

- 

Neither  confirmed 

1  by 

cytology 

+  Total  ED  patients  =  24 

1  of  2  confirmed 

by 

cytology 

ED-CAV  =  13 

ED-CAV/E  =  11 

Table  12  —  Metastatic  Sites  at  Diagnosis  in  Extensive  Disease  (ED) 
Patients  by  Treatment  Arm 


97 


Number  of  patients 


%  of  all  patients  who 


Site 

with  relapse  at  site+ 

who  relapsed  > 

Chest  (excluding  pleura) 

9 

23% 

Liver 

7 

18% 

Bone 

5 

13% 

Brain 

5 

13% 

Nodes  (excluding  chest) 

4 

10% 

Pleura 

2.':- 

5% 

Bone  Marrow 

1 

3% 

Subcutaneous  Nodules 

1 

3% 

Others  CNS  (choroidal) 

1 

3% 

Neither  confirmed  by  cytology;  one  recurrent 


+Total  pts  =  39 


Relapses  at  Sites  of  Initial  Disease'’"' 

Site  Number  of  Relapses 


Liver  4 
Bone  3 
Nodes  (excluding  chest)  2 
Pleura  1 


Excludes  chest  relapses,  except  in  pleura 


Table  13  —  Sites  of  Relapse 


i 


tp 


98 


Response  to  Therapy  by  Stage  and  Treatment  Arm 


All  patients  (n=39)  (no.(%)) 
Limited  Disease  (n=15) 
Extensive  Disease  (n=24) 
CAV-LD  (n=8) 

CAV/E-LD  (n=7) 

CAV-ED  (n=13) 

CAV/E-ED  (n=ll) 


CR 

PR 

NR 

15 

(38%) 

13  (33%) 

11 

(28%) 

12 

(80%) 

1  (  7%) 

2 

(13%) 

3 

(12%) 

12  (50%) 

9 

(38%) 

7 

(88%) 

1  (12%) 

0 

5 

(71%) 

0 

2 

(29%) 

2 

(15%) 

7  (54%) 

4 

(31%) 

1 

(  9%) 

5  (45%) 

5 

(A5%) 

Time  to  Relapse 
Group 
LD-  CR+PR 
ED-  CR+PR 
ED-  NR 

CAV-LD 

CAV/E-LD 

CAV-ED 

CAV/E-ED 


LD  =  Limited-stage  disease 
CR  =  Complete  response 
NR  =  Non-responders 
C  =  cyclophosphamide 
V  =  vincristine 


Median  ( pro  jected) 
361  days 
188  days 
71  days 


p-value 


334  days 
361  days 


.55 


193  days 
109  days 


.04 


ED  =  Extensive-stage  disease 
PR  =  Partial  response 

A  =  Adriamycin  (doxorubicin) 
E  =  Etoposide  (VP-16-213) 


Table  14  —  Treatment  Results;  Response  and  Time  to  Relapse 


1 


Group 

All  patients 
LD  -  CR 


Median  ( pro jected) 


99 


LD-  CR+PR 
ED-  CR+PR 
ED-  NR 

CAV-LD 

CAV/E-LD 

CAV-ED 

CAV/E-ED 


p-value 


301  days 
560  days 


560  days 
230  days 
198  days 


2.  = 
£  = 


.0007 

.24 


560 

424 


.24 


230 

186 


.23 


LD  =  Limited-stage  disease 
CR  =  Complete  response 
NR  =  Non-responders 
C  =  cyclophosphamide 
V  =  vincristine 


ED  =  Extensive-stage  disease 
PR  =  Partial  response 

A  =  Adriamycin  (doxorubicin) 
E  =  Etoposide  (VP-16-213) 


Table  15  —  Survival 


1 


/oo 


L 

L 

L 

L 

L 

LI 

IjJLlILUUJLlI  « 

♦ 

u 

bJ 

♦  • 

♦ 

UJ 

♦  o 

_l 

Ul 

+  o 

• 

-1 

u 

♦  n 

• 

UJ 

UJ 

•  • 

♦  _l 

Ul 

♦o 

_l 

Ul 

-t-uo 

•_J 

Ul 

•  _1 

UIUIUUI 

-I  UJ 

_j  UJUJUUJLlI 

_j  LlIUJUILU 

_l  UJUjUULii 

_j  UiliJUJUl 

-I  u 

-J  llJ 

_j  UJlULdUi 

-I  Ui 

_l  LUUiUJUJ 

_l  UUULJ 
UlUJliJLU 


•  u 

•UJ 

•  UJ 
•Ul 

-t-  Ul 

•  UJ 

•u 

•UJ 

•Ul 

-t-u 

•  -».  .  . 

•o 

+o 

•uo 

•  o 

•  o 

-4- 

o 

o 

o 

o 

o 

o 

o 

o 

o 

o 

uo 

o 

ID 

o 

tn 

o 

in 

o 

o 

m 

n 

o 

rs 

m 

rvi 

o 

* 

00 

rv 

'O 

ID 

n 

n 

• 

• 

♦ 

4 

• 

♦ 

• 

• 

o 

O 

O 

O 


o 

o 


o 

o 

00 


o 

o 

rv 


o 

o 

>0 


o 

o 

b~ 


o 

o 


o 

o 

r>i 


o 

o 


a; 

w 

oj  ro 

W  0) 
03  CO 
QJ  'H 
CO  Q 

•H 

Q  QJ 

> 

T3  -H 
0)  CO 

^  c 

•H  QJ 

E  -i-J 

•H  X 

J  W 

II  II 

J  w 


CO 

CO 

Q 


CD 

E 


C 

OJ 

4-1 

X 

w 

QJ 

CO 

CO 

QJ 

CO 


XI 


Cu 


Di 


CO 

QJ 

"O 

C 

o 

o- 

CO 

QJ 

o; 

u 

o 


QJ 

CO 

COL 

CO 

T— i 

QJ 

Di 


QJ 

E 

•H 

H 

I 


QJ 

U 

3 

00 


asdBxsa  JO  aajj  (juaajaj)  AjTXTqBqojj 


1 


d  d  d  d  d  d  d  d  d  d  d  d  d .  J  d  .-j  d  d  d  d  d  d  d  d  d  d  d  d  d  d  +  000* 


U.U.U.U.U. 


o 

LI 

f  J 

n 

n 

o 

LT 

n 
LT 

rv 

cs 

•n 

♦  o 


u-ZZZZ-t- 


il.  z 

u.  Z 

u.  Z 

n.  z 

.Li.U.il.  Z 

z 

z 

z 

zzzzzz 

z 

z 


o 

C3 

rj 


LT 

r* 


n 

ri 


ii“ 

rv 


o 

o 

rj 


Ce: 

Pm 


a; 

U  cn 
w  u 

0) 

W  T3 

u  c 

OJ  o 

-O  Dh 

c  W 
O  01 
Q.  L- 
cn  C 
<0  O 
2 

II  II 

iX  2 


cn 

ro 

Q 


Z 

Z 

Z 

Z 

Z 

Z 

Z 

z 

z 

z 


-LO 


LP 


z 

zzzzz 

z 

z 

z 

z 

z 

z 


zzzzzz 

z 

z 

z 

z 

z 

z 

z 

z 


zzzzz 

z 

z 

z 

z 

z 

z 

zzzzzz 

zzzzz 


zzzzzz 

z 

z 


z 

z 

zzzz 


o 


•bP 


bP 

o 


o 

bP 


IV 

CD 


o 

o 


o 

bP 


o 

IV 


bP 


o 

o 


o 

bP 


bP 

n 


o 

o 

o 


o 

O 

o 

o 

o 

bP 

o 

bP 

o 

bP 

|V 

bP 

r-i 

o 

IV 

CD 

rv 

>0 

UP 

o 

o 

bP 

r-j 


o 

bP 

rj 


o 

o 

o 


asdB-[aH  Jo  aa-ij  (dusDjaj)  Aru^TQ^qo-id 


Figure  2  —  Time  to  Relapse  for  Extensive  Disease  Patients  by  Response  Group 


1 


.000 


o 

o 

o 


'O 

+liT 


< 

< 

c 

< 

€ 

< 

< 

<  c<.  <:«  c 
<. 
c 
c 
€ 

€ 

c<  c<  « 


1 

i 

CCi  iZiQj 

A 

«  «  «  «  CC  <■<■  <CA  <.<1 
C  A 

«  *==< 

C  AAAAAAiAAA 

«  A 

A  AA  I'^t'*''  ■f"'''- 
A  C 
€ 

< 

< 

< 

<. 

< 

< 

< 

< 

C 


o 

o 

A 


O 

o 


o 

o 

■c 


o 

o 

<r 


•o 

-t-ll*! 

n 


o 

o 

n 


o 

o 

Cm 


o 

+ii‘2 


O 

o 


♦o 


o 

•iiT 


O 

O 


w 

>  > 
<c  < 
u  o 

II  II 
<t;  CQ 


♦  • 

cn 

-o 

>, 

-t-UT 

CO 

♦til 

Q 

. 

c 

♦  O 

•H 

-+-  o 

•  til 

OJ 

E 

•o 

•H 

-*-lll 

•<T 

o 

o 

© 

O 

o 

O 

o 

o 

O 

bl 

o 

111 

o 

111 

o 

N 

m 

fJ 

o 

IS 

U1 

rj 

o 

A 

rv 

vO 

m 

M 

fj 

♦ 

• 

• 

• 

♦ 

• 

• 

o 

asdBX9H  (xuaojaj) 


Figure  3  —  Time  to  Relapse  for  Limited  Disease  Patients  by  Treatment  Group 


io3 


• 

•  in 

• 

•UIM 

“ 

*  • 

♦ 

-t-n 

•  M 

♦ 

CCA  AAA 

•  n 

• 

A 

•  0 

• 

A 

•  • 

• 

A 

UT 

A 

♦ 

A 

•  to 

♦ 

A 

•llT 

♦ 

A 

»  • 

♦ 

A 

-l-fv 

4- 

A 

•  O' 

• 

A 

.r>i 

• 

A 

•  0 

• 

A 

•  * 

* 

AkC«<-«*  0 

A 

♦  m 

*■ 

A 

•  rj 

• 

A 

♦  UT 

♦ 

A 

•  • 

• 

A 

h 

A 

•  >0 

♦ 

A 

•  fi 

• 

A 

-  0 

* 

A 

♦  * 

A 

-t-  liT 

4“ 

A 

*  ^ 

• 

A 

•  rj 

• 

A 

•  U") 

* 

€«4CC« 

< 

c 

< 

* 

I* 

< 

< 

< 

♦ 

c«  <:< 

• 

€ 

€ 

* 

¥ 

* 

cc 

* 

■c 

* 

•c 

c 

ac 

< 

dm 

A  iXiTi  A 

* 

A 

• 

A 

* 

•c 

A 

4- 

< 

A 

A 

A 

■c 

A 

4C 

A 

A 

♦  <: 

A 

•  c 

A 

♦  <. 

A 

♦ « 

A 

4-  <: 

A 

•  < 

A 

*  c 

fff  AA  ObA 

•  c 

A  AA  AA 

•  « 

A 

4-  C 

A 

•  <: 

A 

•  < 

AA  AA 

<  » 

<  » 

<  04 

<  OaiXiiZiiXi 

C  A 
C  A 

<  A 
A 

c  ® 

* 

^  aaaaa 

^  A  AA  AA 

4C  A 

<  A 

C  A 

«  A 

A 
A 
A 
A 
A 
A 
A 
A 
A 
A 
A 
A 
A 
A 
A 
A 

AA  AA 


•  <. 

►  « 
■  « 
•  < 
■•  C 


A 

A 

A 

A 

A 


>  tTjT .  AA  a 


-rv 

►  n 

•  n 


o 

o 


♦  n 

cn 

♦  LD 

>, 

CO 

+  rj 

.0 

Q 

» ^ 

c 

*  0 

•H 

4-  UT 

•  !V 

cu 

• 

E 

*uT 

•H 

+  rv 

• 


O 


•  n 

.  rj 


UT 

O 


•  o 

•U' 

-t-  - 

•  CD 

•  o 

*  o 

♦  o 

♦  rv 

♦  o 

•LT 
+  • 

♦  r-j 
•LT 

•  o 

•  o 


UT 

M 


•O 
■  b~ 


•  A 

4-  A 
-  -t-  • 

*  •4-  • 

.  .  -4-  *  * 

.  .4.  .  .  - 

•4-  •  • 

•  -4- 

000* 

,8750 

0 

0 

LO 

0 

uo 

n 

>0 

.5000 

.3750 

.2500 

.1250 

0 

0 

0 

0 

Ol 

OI 

o 


w 

>  > 
c  c 
CJ  o 

II  II 

C  CQ 


asda^aH  50  aaJj  (:iuaDjaj)  A:iT-[TqBqojj 


Figure  4  —  Time  to  Relapse  for  Extensive  Disease  Patients  by  Treatment  Group 


)((}((]•(  )tc  I(( 


•  o 

-r  C 

*  &- 

.  . 

•O 

*****  -r  J^! 

* 

•  OC' 

« 

•  • 

♦  o 

4-  O 

-  O 

* 

•  • 

•o 

-r-  ij"; 

•  rv 

* 

♦  o 

-r  O 

* 

•  (N 

•  • 

-O 

4-  Si 

* 

.<! 

* 

* 

* 

+  s 

*  'C 

* 

•  • 

* 

* 

* 

*• 

* 

* 

* 

•» 

* 

* 

* 

«• 

*»* 

•*• 

» 

» 

«• 

» 


*** 

* 

*** 

* 

* 

* 

«• 

«• 

*** 

•»* 

** 

*** 

»* 

»*»* 

■» 

** 

* 

* 

«•* 

« 

« 

» 

«• 

* 


o 

o 

o 


o 

o 

<r 


o 

o 


o 

o 


4-  * 


o 

o 


cn 

>1 

cc 

Q 


0) 

E 

•H 

E-h 


o 

o 

O 

o 

o 

o 

o 

O 

o 

o 

LT 

o 

UT 

o 

UT 

o 

UT 

o 

o 

IV 

in 

fj 

o 

r^. 

bl 

r-j 

o 

• 

C3 

rv 

O 

UT 

n 

* 

SuTATAjng  (^uaojaj)  AnxxTqBqojj 


Figure  5  —  Overall  Survival 


000  +EEEEEEELLLLLLLLLLLLLLLLLLLLLLLLLLLLLLLLLLL 


|Ot) 


UiLuUJ  LJUJUJ 


UJ  UJ  LU  LiJ  LU  UJ  LiJ 
UJUJUUJLiJUJ 
llJ 
UJ 
U 
LlI 
UJ 


UIUJUJUJUJUJ 

UlUIUJUiUJ 

UlUJUJUJ 

u 

UJUUUUJUIUI 

UJ 

UlUJUJUJ 

u 

Ui 


Ui 

UI 

u 

UJ 

Ui 

UI 

UI 

UIUJUI 


o 

o 

o 


0) 

♦ 

Cfl 

o 

OJ 

CO 

o 

cn 

OJ 

O' 

CO 

cn 

<u 

•H 

* 

Q 

o 

•H 

yo 

Q 

0) 

00 

> 

TO 

■H 

• 

0) 

CO 

o 

c 

o 

•H 

OJ 

CO 

E 

•H 

X 

* 

w 

o 

II 

lO 

II 

IV 

—1 

u 

o 

o 


o 

o 

>0 


o 

o 

ll" 


o 

o 


o 

o 

Ml 


o 

o 

rj 


o 

o 


o 

o 


o 

o 

o 

o 

o 

o 

o 

o 

in 

o 

in 

o 

m 

o 

m 

o 

IV 

in 

rj 

o 

fv 

uo 

rj 

o 

00 

IV 

>0 

m 

n 

(N 

• 

• 

• 

• 

♦ 

♦ 

• 

• 

o 

CO 

Q 


SuTAiAjng  (:iu03jaj)  X:iT'[TqBqoaj 


Figure  6  Survival  for  Responders  (CR  +  PR)  by  Disease  Extent 


PPPFTPPPFPfP 


o 

c 

o 


o 

L*) 

o 


zz 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 


zzzzzzzzzzz- 


CeC 

o 

Cu 

o 

o- 

+ 

D£ 

o 

u  cn 

CO 

w  U 

0) 

cn  TO 

u  c 

o 

o 

Cu 

OJ  o 

-a  Cu 
c  cn 
O  OJ 

Cl  u 

• 

cn  c 

o 

0)  o 

uD 

cu  2 

Cn 

II  II 

o 

CU  2: 

o 


o 

o 

>0 


c 

li~ 


CD 

Q 


z 

z 

z 


z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 

z 


o 

o 


^u.u.u.u.u.ij. 


o 

o 


o 

Ijl 


rj 


o 

UD 


o 

o 


o 

4iD 


o 

o 


-u  .  . 

o 

o 

O 

o 

o 

o 

o 

o 

o 

o 

UD 

o 

UD 

o 

liD 

o 

yo 

o 

c 

IV 

UD 

fj 

o 

IV 

yo 

r>i 

o 

♦ 

GO 

IV 

>0 

UD 

n 

rj 

• 

♦ 

♦ 

♦ 

♦ 

• 

* 

♦ 

o 

SuTATAjns  (tluaojaj)  X:}T-[iqBqoa j 


Figure  7  —  Survival  for  Extensive  Disease  Patients  by  Response  Group 


lii 


4; 


1 01 


1 


A  >TaTi  iTiA  iTiiTi 

A 


AAA  A 


t-ffi 
A 
A 
A 
A 
t-A 
.A 
A 
A 
.A 
((-A 


O 

o 

o 


•o 

■\n 

•O' 


* 

-  o 

> 

> 

-t-  o 

c 

<c 

♦  O' 

o 

CJ 

•o 

II 

II 

-t-bT 

•A 

< 

CQ 

•  o 

+  o 

-  A 


•o 

•4-liT 

•rv 


o 

o 

(V 


•o 

-hill 

.S3 


O 

O 

>0 


•o 

-UT 


A  A  A  AA  AA  AA  i:c«Xi 

1  *  • 

A 

A 

1  *  ® 

A 

i  +  o 

A 

J  •  bl 

A 

1 

i 

A 

A 

J  •© 

A 

1  +b-3 

A 

* 

A 

»  ^ 

A  <  «« « CC  «€  €« «« C  -  - 

•  AAA  AAAAAAAA  <. 

•  O 

4-A 

< 

•h  O 

.A 

< 

. 

.A 

•C 

. 

.A 

C 

•  • 

.A 

•O 

•rA 

< 

-t-in 

.A 

€ 

•n 

.A 

< 

. 

.A 

< 

o  . 

.A 

C 

♦  o 

4-A 

«  c«  c«  ■€<: «« 

-t-  o 

.A 

•  n 

•  A 

<: 

. 

.A 

< 

.  . 

.A 

•c 

♦o 

l-A 

< 

-HiT 

•  A 

•Tvl 

•  A 

. 

•  A 

.  . 

.A 

< 

♦  O 

•  A 

c 

-t-  o 

.A 

c 

♦  rvi 

•A 

< 

* 

.A 

< 

.  . 

.A 

-o 

•  A 

< 

-HiT 

.A 

< 

.A 

< 

• 

•«X|  «  « 

*  O 

H-O 


O 

o 


o 

o 

o 

o 

o 

o 

o 

o 

O 

o 

b*) 

o 

in 

o 

bT 

o 

bT 

o 

o 

rv 

bT 

n 

o 

rx 

in 

04 

o 

♦ 

ro 

>0 

in 

n 

(M 

♦ 

• 

♦ 

♦ 

• 

• 

♦ 

o 

w 

03 

Q 


SuTAiAjns  (luaojaj)  X^TXTRBqojj 


t 


Figure  8  —  Survival  for  Limited  Disease  Patients  by  Treatment  Group 


vvvvvaa-i-  ooo 


|Og 


♦ro 

< 

• 

c 

•  • 

< 

♦  o 

< 

-H  O 

< 

•  CD 

c 

4b 

c 

•  • 

<: 

*o 

c 

-HbT 

♦r^ 

< 

c 

♦  • 

< 

♦  o 

c 

-H  O 

c 

♦  tv 

•c 

♦O 

< 

-HbT 

•c 

•>0 

•c 

c 

•  ♦ 

« 

-  O 

< 

-H  O 

< 

•  >0 

♦ 

c 

<. 

-o 

< 

-KbD 

< 

-bO 

<. 

♦  • 

•  O 

c 

•H  O 

« 

•  b~ 

c 

< 

c 

♦O 

€ 

-HbD 

< 

•c 

•  * 

■c 

*  O 

< 

+  o 

< 

♦ 

< 

-  • 

c 

•o 

€ 

-HbD 

•c 

-n 

iZaZi  Aa  iXiiZi  AiA  SA 
A  C 

cc  «  c 

AAACCAAAkXAA 
A  <. 


A 

A 

A 

A 


<. 

< 

€ 

•C 


O 

o 

o 


o 

bD 

w 

> 

> 

<c 

<c 

o 

o 

CJ 

Ch 

II 

II 

o 

< 

CQ 

bT 


O 

o 

n 


«< 

<. 

c 

« 

<: 

<: 

« 

c 

«c  « 

A 

A 

A 

A 

AA  AA 


A  < 

•C  «•€  <»  < 

C  C<  A 

<  «  A 

■C  «  <;•€  A 

<  AA  AA  AA  AA 

<  A 
A 
A 
A 
A 

AA  AA  A 

A 

A 

A 

A 

A 

AA  AA  A 
A 

AA  A 


•O 

-♦-bT 

•r-4 


o 

o 

n 


•o 

-HiT 


-HO 


o 

o 

O 

o 

o 

o 

o 

o 

bD 

o 

lO 

o 

bT 

o 

bD 

o 

rv 

bD 

o 

rv 

n 

n 

o 

m 

IV 

>0 

m 

n 

tM 

♦ 

• 

• 

♦ 

♦ 

♦ 

♦ 

o 

CO 

CD 

Q 


o 

o 


o 

o 


SuTATAjns  (3U80j9j)  X:iT -[xqBq o j j 


Figure  9  —  Survival  for  Extensive  Disease  Patients  by  Treatment  Group 


5 


¥ 

I 

I 


4 


bC  w 


:  ;  dical  library 

'  :).:i  u c  r  i  p  t  Theses 


Unpublishel  th' s  submitted  for  the  '''~-3;’,er 
ceposited  in  the  Ya*-e  Reuie  iJ.  Library  ai'e  to  be  usei 
hts  of  the  authors.  BibliographicaJ.  refererces  m 
t  not  be  copied  rri  thou i:  permission  of  the  aut'rors 
being  given  in  subsequent  vritten  or  published  vorh. 


This  thesis  bv 

used  by  the  following  persoas,  whose  signabures  atte 
^bove  restrictions. 


and  Doctor’s 
only  with  due 
ay  be  noted,  bu 
,  and  without  p 


st  their  accept 


NAiiE  MD  ADDRESS