Skip to main content

Full text of "A Stanford professor's career in biochemistry, science politics, and the biotechnology industry : oral history transcript / 2000"

See other formats


University  of  California  •  Berkeley 


Regional  Oral  History  Office  University  of  California 

The  Bancroft  Library  Berkeley,  California 


Program  in  the  History  of  the  Biosciences  and  Biotechnology 


Paul  Berg,  Ph.D. 

A  STANFORD  PROFESSOR'S  CAREER  IN  BIOCHEMISTRY,  SCIENCE  POLITICS, 
AND  THE  BIOTECHNOLOGY  INDUSTRY 


With  Introductions  by 
Daniel  E.  Koshland,  Jr.,  Ph.D. 

and 
Charles  Yanofsky,  Ph.D. 


Interviews  Conducted  by 

Sally  Smith  Hughes,  Ph.D. 

in  1997 


Copyright  ©  2000  by  The  Regents  of  the  University  of  California 


Since  1954  the  Regional  Oral  History  Office  has  been  interviewing  leading 
participants  in  or  well-placed  witnesses  to  major  events  in  the  development  of 
northern  California,  the  West,  and  the  nation.  Oral  history  is  a  method  of 
collecting  historical  information  through  tape-recorded  interviews  between  a 
narrator  with  firsthand  knowledge  of  historically  significant  events  and  a  well- 
informed  interviewer,  with  the  goal  of  preserving  substantive  additions  to  the 
historical  record.  The  tape  recording  is  transcribed,  lightly  edited  for 
continuity  and  clarity,  and  reviewed  by  the  interviewee.  The  corrected 
manuscript  is  indexed,  bound  with  photographs  and  illustrative  materials,  and 
placed  in  The  Bancroft  Library  at  the  University  of  California,  Berkeley,  and  in 
other  research  collections  for  scholarly  use.  Because  it  is  primary  material, 
oral  history  is  not  intended  to  present  the  final,  verified,  or  complete 
narrative  of  events.  It  is  a  spoken  account,  offered  by  the  interviewee  in 
response  to  questioning,  and  as  such  it  is  reflective,  partisan,  deeply  involved, 
and  irreplaceable. 


************************************ 


All  uses  of  this  manuscript  are  covered  by  a  legal  agreement 
between  The  Regents  of  the  University  of  California  and  Paul  Berg 
dated  November  18,  1997.  The  manuscript  is  thereby  made  available 
for  research  purposes.  All  literary  rights  in  the  manuscript, 
including  the  right  to  publish,  are  reserved  to  The  Bancroft  Library 
of  the  University  of  California,  Berkeley.  No  part  of  the 
manuscript  may  be  quoted  for  publication  without  the  written 
permission  of  the  Director  of  The  Bancroft  Library  of  the  University 
of  California,  Berkeley. 

Requests  for  permission  to  quote  for  publication  should  be 
addressed  to  the  Regional  Oral  History  Office,  486  Bancroft  Library, 
Mail  Code  6000,  University  of  California,  Berkeley  94720-6000,  and 
should  include  identification  of  the  specific  passages  to  be  quoted, 
anticipated  use  of  the  passages,  and  identification  of  the  user. 
The  legal  agreement  with  Paul  Berg  requires  that  he  be  notified  of 
the  request  and  allowed  thirty  days  in  which  to  respond. 

It  is  recommended  that  this  oral  history  be  cited  as  follows: 


Paul  Berg,  Ph.D.,  "A  Stanford  Professor's 
Career  in  Biochemistry,  Science  Politics, 
and  the  Biotechnology  Industry,"  an  oral 
history  conducted  in  1997  by  Sally  Smith 
Hughes,  Regional  Oral  History  Office,  The 
Bancroft  Library,  University  of 
California,  Berkeley,  2000. 


Copy  no. 


Paul  Berg,    1980. 


Cataloguing  information 


BERG,  Paul  (b.  1926)  Professor  of  biochemistry 

A  Stanford  Professor's  Career  in  Biochemistry,  Science  Politics,  and  the 
Biotechnology  Industry,   2000,  xiv,  249  pp. 

Childhood  and  education,  New  York  City;  higher  education,  New  York, 
Pennsylvania  State  University,  Washington  University,  St.  Louis; 
postdoctoral  research  with  Arthur  Kornberg  and  Herman  Kalckar;  research  on 
amino  acid  activation,  tumor  viruses,  recombinant  DNA;  Asilomar  I 
conference  (1973),  recombinant  DNA  biohazards  controversy,  Asilomar 
Conference  on  Recombinant  DNA  Molecules  (1975);  commercialization  of 
recombinant  DNA  &  molecular  biology;  Nobel  Prize,  1980;  DNAX  Research 
Institute  of  Molecular  &  Cellular  Biology,  Inc.,  relations  with  Schering- 
Plough;  Beckman  Center  for  Molecular  &  Genetic  Medicine,  Stanford  Medical 
School;  Stanley  N.  Cohen,  Herbert  W.  Boyer,  Peter  Lobban,  James  D.  Watson, 
John  Morrow,  Joshua  Lederberg,  and  other  scientists. 

Introductions  by  Daniel  E.  Koshland,  Jr.,  Professor  of  Molecular  and 
Cell  Biology,  UC  Berkeley,  and  Charles  Yanofsky,  Professor  of 
Biological  Science,  Stanford  University. 

Interviewed  1997  by  Sally  Smith  Hughes  for  the  Program  in  the  History 
of  the  Biological  Sciences  and  Biotechnology,  Regional  Oral  History 
Office,  The  Bancroft  Library,  University  of  California,  Berkeley. 


TABLE  OF  CONTENTS--Paul  Berg 

BIOTECH  SERIES  HISTORY  by  Sally  Smith  Hughes  i 

INTRODUCTION  by  Daniel  E.  Koshland,  Jr.  v 

INTRODUCTION  by  Charles  Yanofsky  vi 

INTERVIEW  HISTORY  by  Sally  Smith  Hughes  x 

BIOGRAPHICAL  INFORMATION  xiv 

I  CHILDHOOD  AND  EDUCATION  1 
Family  and  Early  Education  1 
Abraham  Lincoln  High  School  4 
Sophie  Wolf  5 

II  COLLEGE,  GRADUATE,  AND  POSTGRADUATE  STUDENT  8 
Chemical  Engineering  Student  at  City  College  of  New  York  8 
Biology  Student  at  the  Brooklyn  College  Campus  9 
Biochemistry  Student  at  Pennsylvania  State  University,  1943- 1948    10 
Military  Service  in  World  War  II  11 
Return  to  Penn  State  13 

Marriage  13 

Decision  to  Do  Graduate  Work  in  Biochemistry  13 

Graduate  Student,  Western  Reserve  University,  1948-1952  14 

Applying  to  Graduate  Schools  14 

Decision  to  Attend  Western  Reserve  16 

The  Department  of  Clinical  Biochemistry  16 

Research  on  the  Artificial  Kidney  17 

Joining  the  Department  of  Biochemistry  17 

Research  on  Nutritional  Supplements  19 

Berg's  Initial  Research  Project  20 

Countering  du  Vigneaud  22 

Harland  Wood  and  Radioisotopic  Tracers  22 

Visits  by  Arthur  Kornberg  and  Herman  Kalckar  24 

Postdoctoral  Positions  26 

Deciding  Not  to  Go  to  the  Coris'  Lab  26 
Postdoctoral  Research  Fellow  with  Arthur  Kornberg, 

Washington  University,  1953-1954  27 

Postdoctoral  Research  Fellow  with  Herman  Kalckar,  Institute  of 

Cytophysiology,  Copenhagen,  1952-1953  28 

Discovery  of  Nucleoside  Diphosphokinase  29 

Making  Radioactive  ATP  30 

Life  in  Denmark  31 

James  D.  Watson  32 

More  on  the  Postdoc  with  Kornberg  33 

Institutional  Setting  33 

Lipmann  and  Lynen's  Hypothesis  35 

Berg's  Research  on  Acyl  Adenylates  35 


Rising  Star  38 

Kornberg's  Background  40 

III   RESEARCH  INTERESTS  NEW  AND  CONTINUING  42 

Research  on  Amino  Acid  Activation  42 

Returning  to  a  Curious  Reaction  42 

James  Ofengand's  Research  on  an  Acceptor  43 

Paul  Zemecnik's  Research  on  Amino  Acid  Incorporation  In 

Vitro  43 

Purifying  Enzymes  Activating  Some  Amino  Acids  44 

Mike  Chamberlain  and  Bill  Wood  45 

Arthur  Kornberg's  Research  on  DNA  Replication  46 

Research  on  DNA  Synthesis  46 

Nearest  Neighbor  Experiment  48 

DNA  as  the  Genetic  Material  48 

More  on  Amino  Acid  Assembly  and  Messenger  RNA  Research  49 

Gene  Regulation  51 

Pasteur  Institute  Contributions  51 

Stanford  Biochemistry  Contributions  52 

Dale  Kaiser  and  Bacteriophage  Lambda  53 

Research  on  Tumor  Viruses  54 

Turning  to  Tumor  Viruses  as  an  Experimental  System  54 

Simian  Virus  40  55 

Kornberg's  Dedication  to  E.  coli  as  a  Research  Tool  56 

Choosing  a  New  Research  Direction  58 

Sabbatical  at  the  Salk  Institute,  1967-1968  59 

Establishing  Research  on  SV40  at  Stanford  59 

Building  on  the  Bacteriophage  Lambda-E.  coli  Research  60 

Drawbacks  of  Using  Commercial  Reagents  60 

Biochemical  and  Genetic  Approaches  61 

Fluid  Disciplinary  Boundaries  and  Multidisciplinarity  62 

Research  Leading  to  Recombinant  DNA  Work  63 
Collaborating  with  Charles  Yanofsky  on  Suppressive  Mutations  64 

Using  Phage  as  Transducing  Agents  64 

Developing  a  Transducing  System  for  Mammalian  Cells  65 

Lambda  Bacteriophage  with  Complementary  Tails  66 

Synthesizing  "Sticky  Ends"  67 

DNA  Ligase  67 

Recombinant  DNA  68 

Making  Recombinants  of  SV40  and  Lambda  dv/gal  68 

Peter  Lobban's  Research  on  Recombinant  DNA  68 

The  Jackson,  Symons,  Berg  Paper,  1972  69 

Complexity  of  the  Berg  Recombinant  DNA  Method  70 

The  Cohen-Boyer  Recombinant  DNA  Cloning  Method  70 

Recombinant  DNA  Controversy  71 

Concern  about  Berg's  Proposed  Experiment  of  SV40  71 

Berg's  Involvement  72 

Biosafety  at  Stanford  and  the  Salk  Institute  72 

Asilomar  I  Conference,  1973  73 

The  Moratorium  Letter  and  the  Meeting  at  MIT  74 

Andy  Lewis  and  Natural  Adenovirus-SV40  Recombinants  75 

James  Watson's  Stances  76 


Hypothetical  Risks 

Nonmicrobiologists  and  Research  on  Infectious  Organisms  77 

Achievements  of  the  Research  Moratorium  79 

Transduction  80 

Lysogeny  and  Transduction  80 

Other  Forms  of  Transduction  81 

Transduction  of  Mammalian  Cells  83 

More  on  Recombinant  DNA  Science  86 
Construction  of  Recombinant  DNA  Molecules  to  Study  the 

Mammalian  Cell  86 

Bacteriophage  with  Cohesive  Ends  88 

Creating  Artificial  Cohesive  Ends  88 

Enzymatic  Sealing  of  DNA  Circles  89 

Biochemistry  Department  Contributions  90 

Replication  and  Expression  in  the  Host  Cell  90 

Concern  about  Potential  Biohazards  91 

Janet  Mertz  at  the  Tumor  Virus  Conference  91 

Berg's  and  Lederberg's  Reactions  91 

Putting  the  Experiment  on  Hold  92 

Contributions  to  Recombinant  DNA  Science  94 

David  Jackson's  Opinions  94 

Gobind  Khorana  and  Vittorio  Sgaramella  96 

A  Method  Difficult  to  Execute  98 

Discovery  of  Naturally  Occurring  Cohesive  Ends  98 

Janet  Mertz  98 

Herbert  Boyer  and  Restriction  Enzymes  99 

Mertz  and  Davis:  EcoRl  Makes  Cohesive  Ends  100 

Boyer 's  Group:  Sequencing  the  Cohesive  Ends  101 

First  Experiment  Using  Cohesive  Ends  101 

Peter  Lobban's  Contributions  to  Recombinant  DNA  102 

Thesis  Proposal,  November  1969  102 

Lobban's  Communication  with  the  Berg  Group  103 
Lobban's  Discoveries  and  Speculations  for  Practical 

Applications  103 

Not  Cloning  105 

The  Cohen-Boyer  DNA  Cloning  Experiments  105 

More  on  Recombinant  DNA  106 

More  on  Lobban-Berg  Group  Interactions  106 

Berg  Questions  Lobban's  Use  of  Two  Identical  Molecules  108 

The  Jackson,  Symons,  Berg  Paper,  October  1972  109 

Lobban's  Work  Ignored  109 

Recombinant  DNA:  Jensen,  Wodzinski,  and  Rogoff,  1971  110 

Recombinant  DNA  Construction  Using  Terminal  Transf erase  111 

David  Hogness:  Cloning  of  Eukaryotic  DNA  112 
Discovery  of  Other  Restriction  Enzymes  Making  Cohesive  Ends  113 

Stanley  N.  Cohen  and  the  Cloning  Experiments  113 

Departmental  Affiliation  and  Early  Research  Interest  113 
Mort  Mandel's  Procedure  for  Introducing  DNA  into  Bacteria  114 
Berg's  View  of  the  Genesis  of  the  Cohen-Boyer  Experiments  114 

John  Morrow  and  the  Xenopus  Experiment  114 

Morrow  and  Helling  Challenge  Patent  Inventorship  115 

Berg  Claims  Two  Key  Experiments  in  Cloning  DNA  116 


The  Commercial  Potential  of  Cloning  Technology  116 

Berg  Doesn't  Hear  of  It  116 

Genentech  and  Cetus,  Palo  Alto  117 

Berg's  Nobel  Prize  Address:  Citation  of  Cohen-Boyer  Research  117 

More  on  Berg's  SV40  Experiment:  No  Expectation  of  Cloning  118 

More  on  the  Biohazards  Controversy  119 

MIT  Meeting  to  Discuss  Biohazards,  1974  120 

The  "Berg"  Letter,  July  26,  1974  121 

The  Nobel  Prize  in  Chemistry,  1980  122 

Singling  Out  Berg  122 

Berg's  Opinion  of  His  Best  Work  123 

Speculations  on  Why  Berg  Was  Chosen  124 

Influences  on  the  Choice  of  the  Nobel  Award  127 

Yet  More  on  Recombinant  DNA  128 

Terminal  Transferase  128 

Creating  Permuted  Linear  Molecules  with  "Tails"  128 

Choosing  the  Best  "Tails"  129 

The  Jensen  et  al.  Paper  and  Biochemical  and  Biophysical  Research 

Communications  129 

The  Stanford  Biochemistry  Department's  Industrial  Affiliates 

Program  132 

The  Chemistry's  Industrial  Affiliates  Program  [IAP]  132 

Broaching  the  Idea  of  a  Biochemistry  IAP  133 

Stanford  Relationships  with  Industry  134 

Program  Project  Grant,  Institute  for  Research  on  Aging  134 

Sentiment  Against  lAPs  135 

Functions  of  the  Biochemistry  IAP  135 

Launching  Biochemistry's  IAP  in  the  Late  1970s  137 

Increasing  Commercialization  of  Academic  Biology  137 

Berg's  Prior  Refusal  of  Corporate  Consultantships  137 

Berg  Reconsiders  Corporate  Connections  138 

The  Recombinant  DNA  Controversy  139 

Herbert  Boyer  and  Genentech  139 

William  J.  Rutter  141 

Cetus  Corporation  141 

Stanford's  Policy  on  Consulting  143 

Policy  Reassessment,  1977  143 

The  Shooter  Committee  on  Conflicts  of  Interest  145 

IV    DNAX  RESEARCH  INSTITUTE  OF  MOLECULAR  AND  CELLULAR  BIOLOGY,  INC.    149 

Earlier  Commercial  Ventures  149 

Channing  Robertson's  Company  150 

Alejandro  Zaffaroni  151 

DNAX  152 

Initial  Research  Focus,  Recruitment,  Science  Advisors  152 

Ed  Haber  and  the  Engineering  of  Monoclonal  Antibodies  153 

Utilizing  the  Okayama-Berg  Procedure  154 

Problems  in  Engineering  Monoclonals  155 

Fund-raising  156 

Schering-Plough  and  DNAX  157 

Schering-Plough1 s  Research  History  157 

Scientists'  Initial  Reluctance  158 


J.  Allan  Waltz,  DNAX  President  and  CEO  159 

Tensions  between  DNAX  and  Schering-Plough  159 

ALZA  and  DNAX  161 

DNAX's  Original  Product  Goal  162 

The  Shift  to  T-Cells  163 

The  Expression  Vector  Technique  164 

Recruitment  of  Scientists  164 

Research  Freedom  165 

Cloning  Cytokine  Receptors  166 

A  Long  Discovery  Phase  167 

DNAX  Benefits  Schering-Plough  167 

Arrival  at  Stanford,  1959  169 

Advance  Preparation  169 

An  Unfinished  Science  Building  170 

Settling  In  171 

The  Stanford  Department  of  Genetics  171 

Lederberg's  Arrival  at  Stanford  171 
Stanley  Cohen's  Associations  with  the  Biochemistry 

Department  172 

Lederberg  and  Space  Biology  173 

Faculty  and  Tenor  of  the  Genetics  Department  174 

Minimal  Interaction  between  Biochemistry  and  Genetics  174 

Biochemistry's  Policy  on  Joint  Appointments  175 

Interdisciplinarity  176 

UCSF  176 
A  Consolidated  Stanford  Graduate  Admissions  Policy  in 

Biology  177 

Beckman  Center  Programs  178 

Beckman  Center  for  Molecular  and  Genetic  Medicine  179 

Origin  of  the  Concept  179 

Raising  Funds  180 

Berg's  Strategic  Decisions  182 

The  Program  in  Molecular  and  Genetic  Medicine  184 

Stanford  Biochemistry's  Asilomar  Conferences  185 

Greatest  Contribution  185 

TAPE  GUIDE  188 

SAMPLE  EDITED  PAGE  189 

APPENDIX 

A  Paul  Berg,  CV  and  Publications  190 
B  Selections  from  Berg  papers,  courtesy  Green  Library,  Stanford 

University  208 
C   "The  1980  Nobel  Prize  in  Chemistry,"  Science,  vol.  210,  21 

November  1980  241 

INDEX  244 


BIOTECHNOLOGY  SERIES  HISTORY- -Sally  Smith  Hughes,  Ph.D. 


Genesis  of  the  Program  in  the  History  of  the  Biological  Sciences  and 
Biotechnology 

In  1996,  a  long-held  dream  of  The  Bancroft  Library  came  true  with 
the  launching  of  its  Program  in  the  History  of  the  Biological  Sciences 
and  Biotechnology.   For  years,  Bancroft  had  wished  to  document  the 
history  of  the  biological  sciences  on  the  Berkeley  campus,  particularly 
its  contributions  to  the  development  of  molecular  biology.   Bancroft  has 
strong  holdings  in  the  history  of  the  physical  sciences  —  the  papers  of 
E.O.  Lawrence,  Luis  Alvarez,  Edwin  McMillan,  and  other  campus  figures  in 
physics  and  chemistry,  as  well  as  a  number  of  related  oral  histories. 
These  materials  support  Berkeley's  History  of  Science  faculty,  as  well 
as  scholars  from  across  the  country  and  around  the  world. 

Although  Berkeley  is  located  next  to  the  greatest  concentration  of 
biotechnology  companies  in  the  world,  Bancroft  had  no  coordinated 
program  to  document  the  industry  nor  its  origins  in  academic  biology. 
For  a  decade,  the  staff  of  the  Regional  Oral  History  Office  had  sought 
without  success  to  raise  funds  for  an  oral  history  program  to  record  the 
development  of  the  industry  in  the  San  Francisco  Bay  Area.   When  Charles 
Faulhaber  arrived  in  1995  as  Bancroft's  new  director,  he  immediately 
understood  the  importance  of  establishing  a  Bancroft  program  to  capture 
and  preserve  the  collective  memory  and  papers  of  university  and 
corporate  scientists  and  the  pioneers  who  created  the  biotechnology 
industry.   He  too  saw  the  importance  of  documenting  the  history  of  a 
science  and  industry  which  influence  virtually  every  field  of  the  life 
sciences,  generate  constant  public  interest  and  controversy,  and  raise 
serious  questions  of  public  policy.   Preservation  of  this  history  was 
obviously  vital  for  a  proper  understanding  of  science  and  business  in 
the  late  20th  century. 

Bancroft  was  the  ideal  location  to  launch  such  an  historical 
endeavor.   It  offered  the  combination  of  experienced  oral  history  and 
archival  personnel,  and  technical  resources  to  execute  a  coordinated 
oral  history  and  archival  program.   It  had  an  established  oral  history 
series  in  the  biological  sciences,  an  archival  division  called  the 
History  of  Science  and  Technology  Program,  and  the  expertise  to  develop 
comprehensive  records  management  plans  to  safeguard  the  archives  of 
individuals  and  businesses  making  significant  contributions  to  molecular 
biology  and  biotechnology.   All  that  was  needed  was  funding. 

In  April  1996,  the  dream  became  reality.   Daniel  E.  Koshland,  Jr., 
provided  seed  money  for  a  center  at  the  Bancroft  Library  for  historical 
research  on  the  biological  sciences  and  biotechnology.   Thanks  to  his 
generous  gift,  Bancroft  has  begun  to  build  an  integrated  collection  of 
research  materials—primarily  oral  history  transcripts,  personal  papers, 


ii 

and  archival  collections  —  related  to  the  history  of  the  biological 
sciences  and  biotechnology  in  university  and  industry  settings.   One  of 
the  first  steps  was  to  create  a  board  composed  of  distinguished  figures 
in  academia  and  industry  who  advise  on  the  direction  of  the  oral  history 
and  archival  components.   The  Program's  initial  concentration  is  on  the 
San  Francisco  Bay  Area  and  northern  California.   But  its  ultimate  aim  is 
to  document  the  growth  of  molecular  biology  as  an  independent  field  of 
the  life  sciences,  and  the  subsequent  revolution  which  established 
biotechnology  as  a  key  contribution  of  American  science  and  industry. 

UCSF  Library,  with  its  strong  holdings  in  the  biomedical  sciences, 
is  a  collaborator  on  the  archival  portion  of  the  Program.   David 
Farrell,  Bancroft's  new  curator  of  the  History  of  Science  and 
Technology,  serves  as  liaison.   UCSF  Library  contributed  the  services  of 
Robin  Chandler,  head  of  UCSF  Archives  and  Special  Collections,  who 
carried  out  a  survey  of  corporate  archives  at  local  biotechnology 
companies  and  document  collections  of  Berkeley  and  UCSF  faculty  in  the 
biomolecular  sciences.   The  ultimate  aim  is  to  ensure  that  personal 
papers  and  business  archives  are  collected,  cataloged,  and  made 
available  for  scholarly  research,  both  in  paper  form  and  on  the 
Internet . 

Project  Structure 

With  the  board's  advice,  Sally  Hughes,  a  science  historian  at  the 
Regional  Oral  History  Office,  began  lengthy  interviews  with  Robert 
Swanson,  a  co-founder  and  former  CEO  of  Genentech  in  South  San 
Francisco;  Arthur  Kornberg,  a  Nobel  laureate  at  Stanford;  and  Paul  Berg, 
also  a  Stanford  Nobel  laureate.  A  short  interview  was  conducted  with 
Niels  Reimers  of  the  Stanford  and  UCSF  technology  licensing  offices. 
These  oral  histories  and  others  planned  or  in  progress  build  upon  ones 
conducted  in  the  early  1990s,  under  UCSF  or  Stanford  auspices,  with 
scientists  at  these  two  universitites . '   The  oral  histories  offer  a 
factual,  contextual,  and  vivid  personal  history  that  enriches  the 
archival  collection,  adding  information  that  is  not  usually  present  in 
written  documents.   In  turn,  the  archival  collections  support  and 
provide  depth  to  the  oral  history  narrations. 


1  Hughes  conducted  oral  histories  with  Herbert  Boyer,  William  Rutter,  and 
Keith  Yamamoto  of  UCSF,  and  with  Stanley  Cohen  of  Stanford.   The  first  volume 
of  the  oral  history  with  Dr.  Rutter  is  available  at  the  Bancroft  and  UCSF 
libraries;  transcripts  of  the  other  interviews  are  currently  under  review  by 
the  interviewees. 


iii 

Primary  and  Secondary  Sources 

This  oral  history  program  both  supports  and  is  supported  by  the 
written  documentary  record.   Archival  materials  provide  necessary 
information  for  conducting  the  interviews  and  also  serve  as  essential 
resources  for  researchers  using  the  oral  histories.   The  oral  histories 
orient  scholars  to  key  issues  and  participants.   Such  orientation  is 
particularly  useful  to  a  researcher  faced  with  voluminous,  scattered, 
and  unorganized  primary  sources.   This  two-way  "dialogue"  between  the 
documents  and  the  oral  histories  is  essential  for  valid  historical 
interpretation. 

Beginning  with  the  first  interviews  in  1992,  the  interviewer  has 
conducted  extensive  documentary  research  in  both  primary  and  secondary 
materials.   She  gratefully  acknowledges  the  generosity  of  the  scientists 
who  have  made  their  personal  records  available  to  her:  Paul  Berg, 
Stanley  Cohen,  Arthur  Kornberg,  William  Rutter,  Keith  Yamamoto.   She 
also  thanks  the  archivists  at  the  Bancroft,  UCSF,  and  Stanford 
libraries,  and  personnel  at  Chiron,  Genentech,  and  Stanford's  Office  of 
Technology  Licensing,  for  assistance  in  using  archival  collections. 

Oral  History  Process 

The  oral  history  methodology  used  in  this  program  is  that  of  the 
Regional  Oral  History  office,  founded  in  1954  and  producer  of  over  1,600 
oral  histories.   The  method  consists  of  research  in  primary  and 
secondary  sources;  systematic  recorded  interviews;  transcription,  light 
editing  by  the  interviewer,  and  review  and  approval  by  the  interviewee; 
library  deposition  of  bound  volumes  of  transcripts  with  table  of 
contents,  introduction,  interview  history,  and  index;  cataloging  in 
national  on-line  library  networks  (MELVYL,  RLIN,  and  OCLC) ;  and 
publicity  through  ROHO  news  releases  and  announcements  in  scientific, 
medical,  and  historical  journals  and  newsletters  and  via  the  ROHO  and 
UCSF  Library  web  pages. 

Oral  history  as  an  historical  technique  has  been  faulted  for  its 
reliance  on  the  vagaries  of  memory,  its  distance  from  the  events 
discussed,  and  its  subjectivity.   All  three  criticisms  are  valid;  hence 
the  necessity  for  using  oral  history  documents  in  conjunction  with  other 
sources  in  order  to  reach  a  reasonable  historical  interpretation.1  Yet 
these  acknowledged  weaknesses  of  oral  history,  particularly  its 
subjectivity,  are  also  its  strength.   Often  individual  perspectives 
provide  information  unobtainable  through  more  traditional  sources.   Oral 
history  in  skillful  hands  provides  the  context  in  which  events  occur-- 
the  social,  political,  economic,  and  institutional  forces  which  shape 
the  course  of  events.   It  also  places  a  personal  face  on  history  which 


'  The  three  criticisms  leveled  at  oral  history  also  apply  in  many 
cases  to  other  types  of  documentary  sources. 


iv 

not  only  enlivens  past  events  but  also  helps  to  explain  how  individuals 
affect  historical  developments. 

An  advantage  of  a  series  of  oral  histories  on  a  given  topic,  in 
this  case  molecular  biology  and  biotechnology,  is  that  the  information 
each  contains  is  cumulative  and  interactive.   Through  individual 
accounts,  a  series  can  present  the  complexities  and  interconnections  of 
the  larger  picture.   Thus  the  whole  (the  series)  is  greater  than  the  sum 
of  its  parts  (the  individual  oral  histories),  and  should  be  considered 
as  a  totality. 

Emerging  Themes 

Although  the  oral  history  program  is  still  in  its  infancy,  several 
themes  are  emerging.   One  is  "technology  transfer,"  the  complicated 
process  by  which  scientific  discovery  moves  from  the  university 
laboratory  to  industry  where  it  contributes  to  the  manufacture  of 
commercial  products.   The  oral  histories  show  that  this  trajectory  is 
seldom  a  linear  process,  but  rather  is  influenced  by  institutional  and 
personal  relationships,  financial  and  political  climate,  and  so  on. 

Another  theme  is  the  importance  of  personality  in  the  conduct  of 
science  and  industry.   These  oral  histories  testify  to  the  fact  that  who 
you  are,  what  you  have  and  have  not  achieved,  whom  you  know,  and  how  you 
relate  have  repercussions  for  the  success  or  failure  of  an  enterprise, 
whether  scientific  or  commercial.   Oral  history  is  probably  better  than 
any  other  methodology  for  documenting  these  personal  dimensions  of 
history.   Its  vivid  descriptions  of  personalitites  and  events  not  only 
make  history  vital  and  engaging,  but  also  contribute  to  an  understanding 
of  why  circumstances  occurred  in  the  manner  they  did. 

Molecular  biology  and  biotechnology  are  fields  with  high 
scientific  and  commercial  stakes.   As  one  might  expect,  the  oral 
histories  reveal  the  complex  interweaving  of  scientific,  business, 
social,  and  personal  factors  shaping  these  fields.   The  expectation  is 
that  the  oral  histories  will  serve  as  fertile  ground  for  research  by 
present  and  future  scholars  interested  in  any  number  of  different 
aspects  of  this  rich  and  fascinating  history. 

Location  of  the  Oral  Histories 

Copies  of  the  oral  histories  are  available  at  the  Bancroft,  UCSF, 
and  UCLA  libraries.   They  also  may  be  purchased  at  cost  through  ROHO. 

Sally  Smith  Hughes,  Ph.D. 
Research  Historian 

Regional  Oral  History  Office 
April  1998 


Program  in  the  History  of  the  Biological  Sciences  and  Biotechnology 

Completed  Oral  Histories 

September  2000 


Paul  Berg,  Ph.D.,  A  Stanford  Professor's  Career  in  Biochemistry,  Science 
Politics,  and  the  Biotechnology  Industry,  2000 

Arthur  Kornberg,  M.D.,  Biochemistry  at  Stanford,  Biotechnology  at  DNAX,  1998 

Niels  Reimers,  Stanford's  Office  of  Technology  Licensing  and  the  Cohen/Boyer 
Cloning  Patents,  1998 

William  J.  Rutter,  Ph.D.,  The  Department  of  Biochemistry  and  the  Molecular 

Approach  to  Biomedicine  at  the  University  of  California,  San  Francisco, 
1998 


Oral  Histories  in  Process 


Horace  Barker,  Ph.D. 

Herbert  W.  Boyer,  Ph.D. 

Stanley  N.  Cohen,  M.D. 

Daniel  E.  Koshland,  Ph.D. 

Marian  E.  Koshland,  Ph.D.,  retrospective 

Reorganization  of  Biology  at  UC  Berkeley 

Edward  E.  Penhoet,  Ph.D. 

Robert  A.  Swanson 

Keith  R.  Yamamoto,  Ph.D. 


vi 


INTRODUCTION- -by  Daniel  E.  Koshland,  Jr. 


Paul  Berg  is  one  of  the  giants  of  biochemistry  in  the  twentieth 
century.   He  made  crucial  discoveries  in  the  area  of  intermediary 
metabolism  and  then  went  on  to  be  a  leader  in  the  recombinant  DNA 
revolution.   Not  only  was  he  a  major  player  in  the  science  but  also  had 
a  pioneering  role  in  resolving  the  ethical  and  legal  questions  that  were 
generated  by  the  new  technology. 

At  a  time  of  increasing  controversy,  he  suggested  a  moratorium  to 
allow  scientists  to  digest  the  benefits  and  hazards  of  the  new  vistas  of 
discovery  and  allow  the  public  to  appreciate  the  balance.   That  calm 
moderation  in  the  face  of  escalating  charges  and  counter  charges  led  to 
the  peaceful  resolution  of  the  issue  and  the  guidelines  and  legislation 
that  have  allowed  biotechnology  to  flourish. 

This  oral  history  traces  Paul  Berg's  career  from  his  childhood  in 
Brooklyn,  through  his  days  as  a  superstar  graduate  student  (he  tries  to 
say  it  was  luck,  but  as  Pasteur  pointed  out,  scientific  luck  only 
impacts  on  history  if  it  connects  with  a  prepared  mind),  to  his  days  as 
professor  and  statesman  of  biochemistry  at  Stanford. 

The  advance  of  science  needs  individuals  who  have  the  high 
imagination  to  solve  the  complex  puzzles  of  nature  and  individuals  who 
have  the  wisdom  and  humanity  to  resolve  the  societal  perturbations 
created  by  the  new  discoveries.   Occasionally  it  is  fortunate  to  have 
all  these  qualities  in  one  individual.   This  oral  history  records  the 
events  and  thoughts  of  one  such  rare  individual  who  was  a  major  actor  in 
the  drama  of  the  biological  revolution  of  the  twentieth  century. 

Daniel  E.  Koshland,  Jr. 

Professor  of  Molecular  and  Cell  Biology 

University  of  California,  Berkeley 

Berkeley,  California 
July,  2000 


vii 


INTRODUCTION- -by  Charles  Yanofsky 


Paul  Berg  is  an  exceptional  scientist  and  individual  who  is 
committed  to  improving  everything  that  concerns  him.   As  his  oral 
history  describes,  he  is  responsible  for  numerous  outstanding 
scientific,  educational,  and  administrative  contributions.   Throughout 
his  career  he  identified  important  unsolved  scientific  problems,  and 
then  proceeded  to  provide  the  solutions.   But  he  was  not  content  with 
concepts  alone.   Several  of  his  greatest  achievements  concern  technology 
development  and  improvement.   In  addition  to  his  scientific  advances,  he 
has  made  a  sincere  effort  to  improve  both  the  support  and  understanding 
of  science.   Few  have  accomplished  as  much  of  significance  as  he  has. 
For  his  contributions  to  science  he  has  received  numerous  awards, 
including  the  Nobel  Prize  in  Chemistry. 

I  know  Paul  very  well.   He  and  I  have  been  good  friends  and 
colleagues  at  Stanford  University  for  over  forty  years.   I  have 
witnessed  many  of  his  scientific,  educational,  and  administrative 
contributions.   We  have  practiced  science  and  participated  in  all 
aspects  of  academic  life  in  the  same  university  setting.   Our  families 
have  always  been  very  close  and  have  enjoyed  sharing  numerous  social  and 
other  activities.   In  addition  to  our  interactions  with  one  another, 
Paul  and  I  joined  Alex  Zaffaroni  and  Arthur  Kornberg  in  founding  the 
DNAX  Research  Institute  and  in  guiding  its  research  and  other 
activities  . 

If  I  were  searching  for  a  single  word  to  describe  Paul  based  on  my 
knowledge  of  his  activities  and  achievements,  I  would  choose  the  word 
savvy.   At  scientific  seminars  and  group  meetings,  Paul  listens  intently 
to  everything  that  is  presented.   He  then  asks  thoughtful  and 
intelligent  questions  focused  on  whatever  topic  is  addressed.   In  group 
deliberations  he  is  not  content  until  each  issue  is  dealt  with  fairly, 
properly,  and  thoroughly,  and  at  a  level  reflecting  the  best  thought 
that  can  be  applied.   He  states  his  views  and  explains  his  vision 
clearly  and  emphatically  until  the  wisdom  of  his  position  is  evident  to 
all.   But  he  listens  to  suggestions,  and  has  no  difficulty  modifying  his 
opinions  or  incorporating  the  ideas  of  others  in  his  recommendations. 
He  has  a  very  positive  outlook;  I  am  certain  that  recipients  of  his 
advice  appreciate  his  desire  to  be  helpful. 

Paul  has  played  a  major  role  in  every  enterprise  he  has  become 
concerned  with.   At  Stanford  Medical  School  he  set  the  highest  standard 
as  mentor  and  practicing  researcher.   He  helped  recruit  many  of 
Stanford's  other  "stars,"  and  he  introduced  several  novel  courses  in  our 
teaching  program.   He  also  is  largely  responsible  for  the  presence  on 
campus  of  the  highly  successful  Beckman  Center  for  Molecular  and  Genetic 


viii 

Medicine.   He  has  directed  the  center  since  its  founding  and  has 
introduced  many  innovative  programs  that  position  researchers  at  our 
university  at  the  leading  edge  in  science.   He  has  also  played  a 
significant  role  in  educating  our  country's  politicians  and 
administrators  on  the  value  of  academic  research  and  its  potential 
benefits  to  society.   He  enjoyed  teaching  the  subjects  he  loves, 
emphasizing  their  promise  for  the  future,  and  has  written  several 
excellent  textbooks  that  describe  our  current  knowledge  in  the  areas  of 
biochemistry  and  genetics. 

During  his  career  Paul  has  had  to  cope  with  two  exceptional  more- 
senior  scientists,  Harland  Wood  and  Arthur  Kornberg,  each  of  whom  was 
always  certain  that  his  view  was  right,  regardless  of  the  issue.   Paul 
has  been  close  to  Arthur  throughout  most  of  his  scientific  career  and 
has  learned  how  to  benefit  from  the  genius  of  this  extraordinary  human 
being.   I  suspect  that  there  probably  is  no  subject  that  Paul  and  Arthur 
have  not  discussed  or  argued  about  with  one  another. 

The  scientists  I  know  well  who  have  worked  with  Paul  have  enormous 
respect  and  admiration  for  him.   He  always  had  the  ability  to  think 
beyond  their  immediate  projects  and  could  readily  identify  the  more 
significant  implications  of  their  studies;  routine  research  was  never 
his  objective.   As  you  will  see  in  his  history,  his  research  interests 
changed  with  time,  generally  reflecting  his  desire  to  exploit  new  and 
exciting  developments  in  the  areas  of  science  that  interest  him.   But  he 
could  not  ignore  potential  dangers  from  the  actions  of  the  scientific 
community,  hence  his  active  participation  and  leadership  at  the  Asilomar 
conference  that  recommended  restraints  on  recombinant  DNA  research. 

In  appropriate  situations  Paul  can  be  very  competitive  since  he  is 
always  determined  to  do  his  very  best.   This  was  most  evident  to  me  on 
the  tennis  court;  we  played  tennis  together  regularly  on  weekends  for 
over  forty  years.   We  both  enjoyed  this  diversion  as  an  opportunity  to 
get  our  minds  completely  off  science  and  to  compete  on  even  terms. 
However,  we  could  not  escape  our  love  of  science  and  invariably 
discussed  our  respective  research  programs  with  one  another,  while 
sitting  on  the  bench.   It  was  great  fun  playing  against  him,  or  with 
him,  because  of  his  strong  desire  to  win. 

Paul  enjoys  music,  art,  the  theater,  and  literature,  and  collects 
and  displays  his  preferences  in  modern  art.   Paul's  wife  Millie,  a 
lovely  woman,  understands  his  dedication  to  his  many  activities  and 
gives  him  her  wholehearted,  enthusiastic  support.   But  she  also 
expresses  her  point  of  view  emphatically,  regardless  of  whether  or  not 
she  agrees  with  him.   They  are  fortunate  that  their  son  John  and  his 
family  live  close  by;  they  share  many  interests  and  activities. 

Paul's  lifetime  experiences  undoubtedly  influenced  his  personal 
goals  and  achievements.   But  success  came  to  Paul  Berg  because  he  is 


ix 

very  smart,  and  he  is  determined.   Few  individuals  have  accomplished  as 
much  as  he  has.   We  are  all  fortunate  that  someone  with  his  ability  has 
been  so  dedicated  to  increasing  our  scientific  knowledge  and  improving 
our  quality  of  life. 


Charles  Yanofsky 

Department  of  Biological  Sciences 

Stanford  University 

Stanford,  California 
September  18,  2000 


INTERVIEW  HISTORY--Paul  Berg 


The  subject  of  this  informative  and  revealing  oral  history  will  be 
familiar  to  virtually  anyone  in  biochemistry  and  molecular  biology. 
Even  before  the  Nobel  Prize  in  Chemistry  thrust  him  definitively  onto 
the  scientific  world  stage  in  1980,  Paul  Berg  was  a  prominent  figure. 
He  had  already  established  a  distinguished  record  in  biochemistry  and, 
more  recently,  in  tumor  virus  research.   He  had  also  risen  to 
international  prominence  in  science  politics  as  a  prime  scientific 
spokesman  in  the  controversy  of  the  1970s  over  the  safety  and  regulation 
of  recombinant  DNA  research. 

Professor  Berg  is  an  old  hand  at  giving  an  oral  history.   In  the 
1970s,  he  was  twice  interviewed  for  MIT's  oral  history  project  on  the 
biohazards  controversy,  a  series  noteworthy  because  it  encompasses 
interviews  with  major  figures  in  the  controversy  as  it  unfolded  and 
documents  participants'  initial  ideas  about  the  scientific  and 
commercial  possibilities  of  the  new  technology.   Because  of  this 
previous  coverage,  the  present  oral  history  accentuates  other  aspects  of 
Professor  Berg's  productive  career--his  science,  his  vision  as  director 
of  Stanford's  Beckman  Center  of  Molecular  and  Genetic  Medicine,  and  his 
affiliation  with  DNAX,  a  private  research  institute  (now  owned  by  the 
pharmaceutical  company  Schering-Plough)  which  he  and  Stanford  colleagues 
Arthur  Kornberg  and  Charles  Yanofsky  founded  in  1980.   The  recombinant 
DNA  controversy  is  nonetheless  a  presence  in  this  oral  history,  as  Berg 
reflects  on  how  it  influenced  the  course  of  research  in  the  field  and 
his  nomination  for  the  Nobel  Prize. 

Inevitably,  when  one  rises  to  a  position  of  power  and  prominence 
in  science,  or  any  other  walk  of  life,  controversy  follows.   Berg  of 
course  is  not  immune.   There  has  been  debate  about  the  wisdom  and 
significance  of  the  temporary  moratorium  which  scientists  themselves, 
with  Berg  in  the  lead,  placed  on  certain  kinds  of  recombinant  DNA 
research  in  the  1970s  and  in  their  formulation  of  research  guidelines. 
The  award  of  the  Nobel  Prize,  as  is  so  often  the  case  in  its  century- 
long  history,  has  also  been  questioned  in  terms  of  those  singled  out  for 
the  honor. ;  Berg  describes  in  detail  the  contributions  of  his 
laboratory  to  the  techniques  for  joining  pieces  of  DNA  by  artificial 
cohesive  ends  ("A-T  tailing")  and  how  this  work  relates  to  that  of  Peter 
Lobban,  Vittorio  Sgaramella,  and,  especially  Stanley  N.  Cohen  and 
Herbert  W.  Boyer.   The  reader  will  likely  be  fascinated  by  Berg's 
comments  on  these  and  other  controversial  topics. 


1  In  additon  to  Berg,  the  1980  Nobel  Prize  in  Chemistry  went  to  Frederick 
Sanger  and  Walter  Gilbert  for  the  development  of  DNA  sequencing  techniques. 


xi 

There  are  several  surprises  in  Berg's  narrative.   One  is  that  he 
considers  his  early  research  in  nucleic  acid  and  protein  biochemistry  to 
be  at  least  as  significant  as  his  later  work  with  tumor  viruses  used  as 
probes  to  study  the  structure,  expression,  and  regulation  of  mammalian 
genes . 

A  second  surprise  is  that  several  years  before  the  now-famous 
Asilomar  Conference  of  1975  on  Recombinant  DNA  Molecules,  Berg  was 
sufficiently  concerned  about  the  risk  of  biohazards  arising  from  the 
growing  technical  capacity  to  manipulate  DNA  that  he  organized  a 
conference  on  the  topic.   Most  of  the  participants  in  the  earlier 
conference,  entitled  Biohazards  in  Biological  Research,  Berg  invited  to 
attend  the  later  conference. 

A  third  surprise,  at  least  to  those  not  intimately  familiar  with 
the  history  of  recombinant  DNA  technology,  is  that  Berg  makes  repeatedly 
explicit  that  he  makes  no  claim  to  the  development  of  molecular  cloning, 
an  achievement  which  he  openly  concedes  to  Stanley  Cohen  and  Herbert 
Boyer.   Although  Gobind  Khorana  and  others  had  previously  joined  DNA 
molecules  synthetically,  Berg  claims  for  his  own  laboratory  the 
development  of  technology  for  using  mammalian  viruses  to  carry  foreign 
genes  into  animal  cells.   The  Berg  group  used  this  "gene-splicing" 
technology  from  1972  on  to  study  the  dauntingly  complex  structure  and 
function  of  mammalian  genes.   Berg  goes  on  to  describe  the  genesis  of 
the  Beckman  Center  for  Molecular  and  Genetic  Medicine  and  its  goal  of 
generating  biomedical  knowledge  which  can  be  translated  into  clinical 
application. 

Berg's  demanding  and  sometimes  contentious  career  has  in  no  way 
diminished  his  energetic  style  and  enthusiasm  for  a  diversity  of 
interests.   To  this  writer,  he  comes  across  as  friendly,  upbeat,  and 
very  likeable.   He  is  also  self-confident,  assertive,  and  articulate. 
He  is  widely  known  in  his  public  and  private  lives  as  a  concerned 
statesman  of  science  and  a  socially  responsible  citizen. 


The  Oral  History  Process 

When  I  approached  Professor  Berg  in  late  1997  about  conducting  an 
oral  history,  he  expressed  concern  about  taking  time  away  from  his 
project  at  Stanford's  Center  for  Advanced  Study  in  the  Behavioral 
Sciences  to  write  a  biography,  with  colleague  and  friend  Maxine  Singer, 
of  the  biochemist  George  Beadle.   He  agreed  to  an  oral  history  with  the 
proviso  that  the  interviews  be  limited  to  four.  Although  we  held  to 


xii 

that  number,  the  duration  of  each  interview  expanded  as  Berg  became 
increasingly  engaged  in  telling  his  story.   My  previous  research  for 
interviews  with  Arthur  Kornberg  and  the  review  I  made  of  the  rich 
collection  of  Berg's  correspondence,  both  collections  archived  in 
Stanford's  Green  Library,  provided  institutional  context  and  a  basis  for 
questions.   In  the  discussion  of  his  research  contributions,  Berg  the 
educator  and  translator  of  science  shines  through  in  the  clarity  and 
completeness  of  his  answers,  including  the  diagrams  of  enzymatic 
reactions  he  created  as  he  reviewed  the  transcripts.   His  opinion  that 
his  greatest  contribution  is  to  have  taught  several  generations  of  young 
scientists  indicates  the  centrality  of  teaching  in  his  lengthy  list  of 
accomplishments . 

Although  an  oral  history  is  assuredly  not  the  best  means  for 
determining  the  factual  content  of  a  scientist's  research—published 
papers,  lectures  and  so  on  are  a  far  better  source--Berg' s  clear  and 
detailed  description  of  his  somewhat  arcane  research  is  accessible  to 
readers  with  only  a  smattering  of  biological  knowledge.   The  four 
interviews  were  conducted  at  Stanford  between  July  15  and  November  5, 
1997,  the  first  two  in  Berg's  office  in  the  Beckman  Center.   The  last 
two  sessions  were  taped  in  his  study  at  the  Center  for  Advanced  Study, 
against  a  background  of  the  raucous  cries  of  California  blue  jays  in  the 
oaks  dotting  the  bucolic  setting  above  the  Stanford  campus.   The  two 
final  interviews  were  interrupted  for  lunch  on  the  center's  sunny  patio, 
where  fellows  are  expected  to  gather  once  a  day  for  scholarly  and  social 
exchange.   Berg  was  characteristically  disturbed  —  an  indication  of  his 
sense  of  social  responsibility—that  myriad  duties  had  kept  him  from  the 
center  more  often  than  he  had  wished. 

The  lightly  edited  transcripts  were  sent  to  Berg  who  painstakingly 
edited  and  amended  them,  despite  his  distaste  for  the  task.   In  a  letter 
accompanying  the  returned  transcripts  (a  sample  of  his  editing  may  be 
found  in  the  appendix),  he  remarked:  "I  thought  the  ordeal  would  never 
end  but  it  has,  I'm  sure,  much  to  your  relief.   Sitting  those  many  hours 
with  you  was  a  pleasure  which  is  more  than  I  can  say  about  the  many  more 
hours  spent  reading  the  transcript:  nearly  three  hundred  [double-spaced] 
pages  was  almost  more  than  I  or  anyone  should  be  expected  to  bear."  The 
fact  that  he  not  only  bore  but  considerably  improved  the  content  and 
clarity  of  his  narrative  is  a  gift  for  which  I  am,  and  future  scholars 
will  be,  highly  grateful  and  appreciative. 

In  1998,  Berg  retired  from  his  professorship  but  continues  to 
direct  the  Center  for  Molecular  and  Genetic  Medicine  and  to  be  an  active 
force  in  American  and  international  science  and  politics. 

The  Regional  Oral  History  Office  was  established  in  1954  to 
augment  through  tape-recorded  memoirs  the  Library's  materials  on  the 
history  of  California  and  the  West.   Copies  of  all  interviews  are 
available  for  research  use  in  The  Bancroft  Library  and  in  the  UCLA 


xiii 

Department  of  Special  Collections.   The  office  is  under  the  direction  of 
Willa  K.  Baura,  Division  Head,  and  the  administrative  direction  of 
Charles  B.  Faulhaber,  James  D.  Hart  Director  of  The  Bancroft  Library, 
University  of  California,  Berkeley. 


Sally  Smith  Hughes,  Ph.D. 

Research  Historian  and  Program  Director 
July  19,  2000 

Regional  Oral  History  Office 
The  Bancroft  Library 
University  of  California,  Berkeley 


xiv 


Regional  Oral  History  Office 
Room  486  The  Bancroft  Library 


University  of  California 
Berkeley,  California  9A720 


BIOGRAPHICAL  INFORMATION 
(Please  write  clearly.   Use  black  ink.) 


Your  full  name 


Date  of  birth 


*  $D  I 


16 


Father's  full  name 
Occupation 


Mother's  full  name 

Occupation 

Your   spouse 


Occupation 


l\f 


Your  children 


Birthplace 


Birthplace 


Birthplace 


Birthplace 


- 
\  J"Ur» 


.     -  U/A 


K?£J'< 


~> 
s>  L,    ^  j 


Where   did   you   grow  up? 
Present    community 


Qy  c.          «•  ft        >^  y. 
'v7\^'*       I  .<5 


Education 


fc-v*  frfrAf    J/U    (.KVg       fy  0     kj^^>^  v  '•'  •»•  - 


Occupation(s ) 


Areas  of  expertise 


T 


' 


Other  interests  or  activities 


I 
j^      fcl    X_ 


Organizations  in  which  you  are  active  _  \f 


V       I   -,. 

>    ^^ 


INTERVIEW  WITH  PAUL  BERG,  PH.D. 


I   CHILDHOOD  AND  EDUCATION 

[Interview  1:  July  15,  1997]«' 

Family  and  Early  Education 

Hughes:   Let's  start  with  your  birth  and  upbringing. 

Berg:     I  was  born  in  Brooklyn,  New  York,  on  June  30,  1926.   My  mother, 

Sarah  Brodsky,  tells  me,  a  very,  very  scorching  day.   The  reason  I 
happen  to  remember  is  because  she  raises  it  each  time  we  have  an 
anniversary.   It  happened  on  June  30,  she  will  say,  "just  like  the 
day  you  were  born." 

My  parents  had  immigrated  from  Russia,  and  under  rather 
unusual  circumstances.   I  think  it  was  1919  that  they  married.   My 
mother  was  eighteen;  my  father,  Harry  Berg,  was  nineteen,  and  they 
left  the  very  next  morning,  never  to  see  anybody  in  their  families 
again.   They  literally  worked  their  way  across  Europe  over  a 
period  of  three  years.   They  had  a  child  and  eventually  came  to 
the  United  States  in  1922,  I  guess  it  was,  arriving  in  New  York 
and  then  settling  in  New  York. 

Hughes:   Was  New  York  their  destiny  when  they  started  out? 

Berg:     I  think  so,  because  there  were  some  members  of  the  family,  a  half 
sister,  who  was  living  already  in  New  York  City.   And  there  must 
have  been  other  people  who  had  come  earlier  from  the  same  little 
town  that  they  were  from,  a  village  outside  of  Minsk.   So  they 
formed  a  local  community  that  knew  each  other  from  Russia,  or  that 
knew  of  each  other  from  Russia. 


'////  This  symbol  indicates  that  a  tape  or  tape  segment  has  begun  or 
ended.   A  guide  to  the  tapes  follows  the  transcript. 


My  father,  I  think,  worked  for  other  people  for  a  while,  and 
ultimately  started  his  own  little  business,  which  was  making  fur 
trimming  on  coats  and  collars  and  hats  and  things  of  that  sort. 
The  boy  that  they  had  while  traveling  in  Europe  died  not  long 
after  they  arrived  in  this  country,  of  what,  I  don't  remember. 
And  so  I  was  born  some  four  years  after  they  arrived. 

We  lived  in  Brooklyn  and  I  can't  remember  a  heck  of  a  lot 
about  my  early  childhood.   I  was  told  that  I  didn't  speak  English 
until  I  went  to  school,  that  I  spoke  mostly  Yiddish,  which  is  what 
my  parents  spoke  to  each  other.   But  once  I  went  to  school,  I  very 
quickly  almost  lost—well,  I  didn't  lose  the  ability  to  understand 
Yiddish,  although  I  lost  the  ability  to  speak  it.  We  lived  in 
what  is  now  the  Brownsville  section  of  Brooklyn,  a  pretty  tough 
neighborhood;  at  that  time  it  was  still  reasonably  nice. 

I  went  through  elementary  school,  probably  through  the 
fourth  grade,  in  that  location.   I  can't  remember  very  much  about 
school.   It  was  certainly  not  challenging  or  that  I  felt  strapped. 
It  was  easy;  I  enjoyed  it;  it  was  fun.   And  then  we  moved  to  a 
place  in  Brooklyn  called  Sea  Gate. 

Sea  Gate  is  a  small  community,  which  at  one  time  was  very 
exclusive  and  very  private.   It's  out  at  the  very  end  of  the 
peninsula  which  forms  Coney  Island.   There's  a  little  peninsula 
that  comes  out  from  the  southern  part  of  Brooklyn,  the  burrough, 
and  out  at  the  very  tip  is  this  little  enclosure  called  Sea  Gate. 
In  the  1920' s,  it  was  probably  quite  an  exclusive  resort,  a  summer 
place  for  wealthy  people  to  come  to,  because  the  homes  were  very 
large. 

By  the  time  we  moved  there,  those  homes  had  become  more 
rooming  houses,  and  it  was  nothing  very  special,  other  than  a 
great  place  to  grow  up.   During  the  wintertime  it  was  literally 
empty.   So  it  was  a  very  small  community  of  young  people  that  I 
knew  and  my  family  knew.   In  the  summertime  it  was  inundated  by 
summer  holiday  people.   But  the  beaches  were  right  there,  so  we 
literally  grew  up  at  the  seashore.   I  think  we  moved  there  before 
I  reached  the  fifth  grade,  because  I  think  I  was  nine  or  ten  when 
I  moved  there.   So  that  was  probably  somewhere  around  the  fourth 
grade. 

Hughes:   Did  that  move  represent  a  rise  in  the  family  fortunes? 

Berg:    No.   I  can't  really  remember  exactly.  We  went  there  for  the 

summer,  again  as  one  of  the  summer  crowd,  and  I  guess  my  father 
and  mother  liked  it  so  much,  they  decided  to  move  there.   We 
didn't  have  a  house;  we  rented  an  apartment,  and  we  lived  in  that 
apartment.   I  then  finished  elementary  school  in  Coney  Island. 


There  were  no  schools  in  Sea  Gate,  so  you  had  to  walk;  it  was 
probably  half  a  mile,  something  of  that  length. 

I  graduated  from  that  school  and  started  junior  high  school. 
Today,  it's  called  middle  school,  I  guess.   It  was  the  seventh, 
eighth,  and  ninth  grades.   I  had  done  very  well  in  school;  I  had 
skipped  grades  twice  during  this  period.   I  guess  that's  not 
common  anymore.   I  forget  which  ones  I  skipped,  but  by  the  time  I 
was  in  the  sixth  grade  and  went  to  junior  high  school,  I  was 
already  a  year  ahead.   There  I  entered  rapid  advance  classes, 
which  took  the  seventh  and  eighth  grade  in  one  year.   So  you  took 
7A  and  7B  in  one  half  year,  and  8A  and  8B  the  second  half  of  the 
year. 

That  was  one  of  the  exhilarating  periods  of  my  life.   The 
people  who  were  selected  to  participate  in  these  rapid  advance 
classes  were  all  very  bright  and  very  energetic,  and  so  it  was  an 
exceedingly  exhilarating  period.   Because  the  teachers  didn't  have 
to  worry  about  people  who  were  slow  in  picking  up  on  things,  you 
could  do  things  in  that  class  that  you  couldn't  do  in  ordinary 
classes . 

Hughes:   Do  you  remember  being  interested  in  anything  in  particular? 

Berg:    I  was  interested  in  science  right  from  the  beginning.   Even  before 
junior  high  school,  I  was  interested  primarily  in  biology.   I  was 
interested  in  trying  to  understand  living  things.   Every  time  I 
found  any  kind  of  an  animal  that  had  died,  I  took  it  home  to 
dissect  it  and  see  what  I  could  learn  about  it.   I  knew  that 
biology  was  the  focus;  that  was  the  thing  I  wanted.   Probably  at 
that  early  stage,  it  was  translated  into  an  ambition  to  be  a 
doctor. 

From  the  things  we  read  during  this  junior  high  school 
experience,  there  was  a  lot  of  motivation  and  idealization  of 
being  a  physician,  or  a  research  physician  in  particular.   I  ask 
my  students  today  how  many  have  read  Sinclair  Lewis's  Arrowsmith 
or  [Paul]  de  Kruif's  book,  Microbe  Hunters.   None  of  my  students 
have  ever  heard  of  either  one  of  them,  which  is  a  disappointment. 
But  the  key  figures  that  were  created  or  converted  into  idols  were 
physicians  who  were  doing  research  and  solving  major  health 
problems.   So  I  had  in  mind  that  I  probably  wanted  to  be  a  doctor, 
but  I  didn't  necessarily  think  of  it  as  practicing  medicine  as 
much  as  doing  research  in  medicine. 

Hughes:   What  did  your  parents  say  about  all  this? 

Berg:    My  parents  had  never  had  any  formal  schooling.   They  left  this 
small  town  before  they  had  any  significant  amount  of  schooling, 


other  then  probably  just  grade  schooling.   They  could  read  and 
write.   But  there  was  nothing  but  encouragement,  nothing  but 
strokes,  lots  of  admiration.   They  knew  I  was  doing  well.   They 
never  ever  queried  me  about  what  I  wanted  to  be  or  anything  like 
that.   They  just  took  great  pleasure  in  having  their  son  being 
successful  in  something  which  they  regarded  as  very,  very 
important—education  at  a  very  high  level—even  though  they  never 
had  it. 

I  think  that  was  true  of  many  of  the  immigrant  Jewish 
families.   Education  was  put  on  a  very  high  level,  even  though  the 
parents  had  little  of  it  themselves.  My  wife,  Mildred  Levy,  says 
today,  because  she  met  my  parents  when  we  were  married,  "You  were 
so  fortunate,  because  your  parents  not  only  encouraged  you,  but 
they  gave  you  enormous  positive  feedback.   So  all  the  while  you 
were  extremely  secure  in  what  you  were  doing,  feeling  good  about 
it.   Without  their  bragging  about  it  or  anything  like  that,  you 
just  knew  you  were  doing  well.   Your  parents  thought  well  of  you, 
and  that  was  great.   You  didn't  have  to  impress  them." 


Abraham  Lincoln  High  School 


Berg:    At  the  same  time,  I  was  very  active  in  sports  and  very  much 

involved  in  playing  football.   Whatever  the  sport  of  the  season 
was,  I  was  actively  involved.   And  so  when  1  went  to  high  school, 
I  really  wanted  to  play  football  as  well  as  do  what  I  had  to  do  in 
school.   But  because  I  had  skipped  so  much,  I  was  really  much 
younger  than  people  who  were  at  the  equivalent  level  in  school,  so 
I  wasn't  as  fully  physically  developed.   I  think  I  was  undersized 
and  would  probably  have  gotten  killed  if  I  went  out  to  play 
football. 

Hughes:   You  were  how  old? 

Berg:     I  graduated  in  January  of  1943.   So  I  went  to  high  school  three 
years  earlier  than  that.   That's  1940,  so  I  was  not  quite 
fourteen.   And  high  school  was  fun.   I  didn't  take  it  seriously. 
I  wasn't  intensely  focused  on  books.   I  just  found  it  easy,  and  I 
could  do  lots  of  other  things. 

But  all  that  time  I  was  very  interested  in  biology.   The 
biology  course  was  one  of  the  really  exciting  and  interesting 
things  we  did  in  junior  high.  We  did  a  lot  biology.   Because  of 
the  quality  of  the  students,  we  did  all  kinds  of  projects. 

Hughes:   What  about  the  quality  of  the  teachers? 


Berg:    My  recollection  is  that  the  teacher  that  we  had  in  junior  high 

school  for  the  rapid  advance  classes  was  magnificent.   She  really 
knew  how  to  handle  gifted  kids.   She  didn't  hold  us  back.   She 
encouraged  us.   1  don't  remember  so  much  about  her,  or  him,  I'm 
not  even  sure  which  one  it  was. 

The  teacher  in  high  school  had  a  very  different  style  of 
encouraging  young  people.   We  were  given  lots  of  projects  to  do, 
and  the  projects  were  to  be  done  at  your  initiative,  and  you 
weren't  given  a  lot  of  help.   There  was  an  enormous  amount  of 
interchange  amongst  the  students  in  the  class,  and  we  all  knew 
each  other.   Many  of  us  were  growing  up  in  the  same  little 
community.   We  always  regarded  ourselves  as  a  special  group, 
because  we  lived  together  during  the  winter  when  nobody  else  was 
there.   And  we  were  the  "in"  crowd,  the  real  crowd. 

During  the  time  I  was  growing  up,  we  had  a  football  team,  on 
which  I  played.   We  played  in  a  league,  and  we  played  tournaments. 
My  brother,  Jack,  who  is  a  year  and  half  younger  than  I  am,  was 
also  on  the  football  team,  and  also  was  a  very  good  student.   We 
were  very  close. 

Hughes:   Just  the  two  of  you? 

Berg:  We  had  a  third  brother,  Irving,  who  was  five  years  behind  me.  At 
that  age,  I  think,  we  had  nothing  to  do  with  him.  There  was  just 
Jack  and  myself. 

When  I  went  to  high  school,  I  took  all  the  science  courses 
that  were  available.   But,  again,  I  don't  remember  knocking  myself 
out;  I  certainly  wasn't  an  honor  student;  my  name  isn't  on  the 
board  as  one  of  the  great  students  —  like  Arthur  Kornberg's  name  is 
there.1   We  went  to  the  same  high  school.   I  couldn't  play  contact 
sports,  because  I  was  not  up  to  that.   So  I  just  played  the  kinds 
of  sports  we  did  at  home. 


Sophie  Wolf 


Berg:    There  was  a  woman,  Sophie  Wolf,  who  was  probably  one  of  the  more 
important  figures  in  my  life  in  terms  of  motivation.   She  was  not 
a  teacher,  but  she  ran  the  supply  room  for  providing  the 


1  See  Arthur  Kornberg,  Biochemistry  at  Stanford  and  Biotechnology  at 
DNAX,  Regional  Oral  History  Office,  The  Bancroft  Library,  University  of 
California,  Berkeley,  1998. 


demonstrations  and  the  microscopes  for  biology  and  the  models  for 
various  lectures.   There  was  this  very  large  stockroom,  which  had 
an  enormous  amount  of  stored  resource  material—microscopes ,  and 
models  of  different  organisms,  and  things  of  that  sort. 

She  had  a  keen  interest  in  young  people.   She  ran  what  was 
called  the  Biology  Club,  which  met  in  people's  spare  time,  after 
classes  were  over.  And  so  1  used  to  spend  the  afternoons  there  at 
the  Biology  Club.   One  of  the  terrific  things  about  her  was,  she 
never  gave  you  any  answers,  no  matter  what  you  asked  her.   She 
would  come  back  with  a  leading  question  that  would  perhaps  help 
you  go  find  out  on  your  own.   This  even  included  doing 
experiments,  so  that  if  you  asked  a  question  which  she  thought  you 
could  answer  by  actually  doing  an  experiment,  she  would  help  you 
to  find  out  what  kind  of  an  experiment  you  might  do.   But  in  the 
end,  it  was  your  drive  that  led  you  to  do  those  kinds  of 
experiments.   I  think  during  the  two  or  three  years  that  I  was  in 
involved  in  this  club,  she  was  constantly  stimulating  us  with 
questions  and  leading  us  on  to  trying  to  learn  new  things. 

Interestingly  enough,  Sophie  was  also  there  at  the  time 
Kornberg  was  a  high  school  student.   At  the  time  I  got  the  Nobel 
Prize  [1980],  somebody  asked  me  if  there  was  any  notable  person 
who  played  a  key  role  in  stimulating  me,  and  I  mentioned  Sophie. 
I  thought  she  was  long  since  dead,  but  as  it  turned  out  she  was 
retired  and  living  in  Florida.   The  education  reporter  for  the  New 
York  Times  wrote  a  very  long  article,  because  it  was  unusual  for 
anybody  to  identify  a  high  school  -figure  as  being  such  a  key 
person.   Usually,  people  identify  a  university  professor  who 
played  a  key  role  in  moving  them  along.   She  then  literally  came 
out  of  anonymity  and  became  quite  well  known. 

Subsequently,  a  third  person  from  Abraham  Lincoln  High 
School  got  the  Nobel  Prize,  in  addition  to  Kornberg  who  received 
the  prize  in  1959,  a  man  by  the  name  of  Jerome  Karle,  who  did  some 
very  magnificent  work  in  crystallography.   So  some  time  ago,  I 
forget  exactly  what  year  it  was,  they  decided  to  name  the  science 
wing  of  the  high  school—it  was  a  very  large  high  school—the 
Sophie  Wolf  Wing.   And  one  floor  is  named  the  Paul  Berg  floor,  and 
the  other  one  is  Arthur  Kornberg,  and  the  other  is  Jerome  Karle. 

They  had  this  huge  assembly;  all  the  students  were  gathered 
in  this  large  auditorium,  and  the  mayor  and  the  secretary  for 
education,  or  whatever  it  was,  for  the  City  of  New  York  came.  And 
there  was  little,  old,  wiry,  Sophie  Wolf,  who  was  pretty  far  out 
of  it  most  of  the  time,  but  she  just  lapped  up  the  accolades. 

Hughes:   There  can't  be  too  many  individuals  at  the  high  school  level  who 
are  able  to  boast  of  mentoring  three  Nobel  prize  winners. 


Berg:    I  don't  know  if  it  was  the  only  high  school  in  the  U.S.  that  has 
had  three  Nobel  graduates,  maybe  the  Bronx  High  School  of  Science 
or  Clinton  High  School,  which  were  also  very  academically 
oriented.   But  my  school  was  not  specially  notable  for  its 
academic  achievements.   It  was  big.   It  was  located  in  an  area 
called  Brighton  Beach,  which  was  quite  a  way  from  where  I  lived. 
We  needed  a  bicycle,  or  in  the  wintertime  there  was  a  bus  that 
took  us,  and  sometimes  we  even  walked.   It  was  probably  three  to 
four  miles. 

Hughes:   How  did  Sophie  Wolf  get  her  scientific  knowledge? 

Berg:    I  think  just  on  the  job.   I  don't  know  how  she  got  into  that  role. 

The  high  school  opened  in  1930.   I  think  Kornberg  moved  to 
it  from  another  high  school  because  it  was  closer  to  his  home. 
And  1  think  he  only  went  there  the  final  two  years  of  his  high 
school  stay.   He  graduated  high  school  at  a  very  young  age.   I 
think  he  graduated  from  City  College  when  he  was  sixteen.   So, 
it's  conceivable  that  he  went  to  high  school  when  he  was  twelve  or 
thirteen. 

But  in  any  case,  Arthur  acknowledges  the  impact  that  Sophie 
Wolf  had.   Jerome  Karle  did  as  well.   She  not  only  was  for 
biology,  but  she  managed  the  storeroom  for  all  the  demonstrations 
for  physics,  chemistry,  and  so  on.   I  don't  know  whether  she  had 
any  science  background  at  all.   She  must  have  been  there  right 
from  the  beginning  when  the  school  opened.   But  it  was  a  central 
role,  because  she  saw  all  the  science  students.   And  for  anybody 
who  expressed  an  interest,  she  would  invite  them  to  join  one  or 
another  of  the  clubs  that  she  had. 

Hughes:   It  had  to  be  by  her  invitation? 

Berg:    Well,  either  an  invitation  or  encouragement,  one  or  the  other. 

Anyway,  while  I  was  in  high  school,  Pearl  Harbor  occurred. 
I  graduated  in  1943,  and  I  remember  very  clearly  the  big  assembly. 
The  school  was  assembled  in  this  large  auditorium  to  listen  to 
President  Roosevelt  condemn  this  "act  of  infamy"  and  then  declare 
war.   So  my  last  year  or  year  and  a  half  in  high  school,  we  were 
very  much  taken  up  with  the  war,  and  what  we  were  going  to  do,  and 
how  we  were  going  to  participate.   I  had  made  up  my  mind  that  as 
soon  as  I  turned  of  age,  I  was  going  to  enlist  in  the  navy.   I 
graduated  in  1943,  so  I  was  sixteen  and  a  half.  Almost 
immediately  after  I  graduated,  a  friend  of  mine  and  I  enlisted  in 
the  navy  air  corps,  the  navy  flight  program,  and  we  were  to  be 
called  up  somewhere. 


II   COLLEGE,  GRADUATE,  AND  POSTGRADUATE  STUDENT 


Chemical  Engineering  Student  at  City  College  of  New  York 


Berg:    In  the  meantime,  I  certainly  had  the  ambition  to  go  to  college. 
My  family  wasn't  well  off  enough  to  allow  me  to  go  wherever  I 
wanted,  so  I  went  to  City  College  in  New  York,  which  was  free  if 
you  had  the  appropriate  grades.   And  so  I  was  admitted  to  City 
College.   Now  I  can't  understand  why,  but  at  that  time  I  set  out 
to  be  a  chemical  engineer.   Quite  different  than  all  this  medical 
ambition  I  grew  up  with.   I  don't  know  who  prompted  me  to  think 
that  going  into  medicine  was  difficult  for  a  Jewish  guy  at  that 
stage.   I  know  Kornberg  himself  had  the  same  reservations,  the 
same  concerns,  and  the  same  warnings.1  Chemical  engineering  was 
something  that  was  practical,  was  something  that  you  could  easily 
count  on  as  a  career  and  for  making  a  living. 

Anyway,  that  whole  thing  at  City  College  lasted  three  days. 
Because  from  where  I  lived,  which  was  at  the  southern  tip  of 
Brooklyn,  it  took  me,  to  go  to  City  College  in  upper  Manhattan, 
almost  three  hours  to  commute  one  way,  two  and  a  half  the  other. 
I  was  committed  to  being  a  chemical  engineer,  but  I  had  already 
enlisted  in  the  navy,  and  I  knew  that  they  were  going  to  call  me 
sometime. 

The  procedure  at  City  College  for  registering  for  classes 
was  about  as  baroque  as  anything  you  could  possibly  imagine. 
There  was  a  certain  number  of  courses  that  were  available.   And  in 
one  room  of  this  immense  hall,  they  had  all  the  courses  listed  up 
on  a  board.   As  soon  as  a  sufficient  number  of  students  had 
registered  for  that  course,  they  took  it  off  the  board.   You  tried 


1  For  Kornberg 's  view  on  anti-Semitism,  see  Arthur  Kornberg, 
Biochemistry  at  Stanford  and  Biotechnology  at  DNAX,  Regional  Oral  History 
Office,  The  Bancroft  Library,  University  of  California,  Berkeley,  1998. 


to  make  up  your  schedule  sitting  in  a  room  three  hundred  yards 
away  from  a  printed  schedule.  After  you  made  up  a  schedule,  you 
got  in  a  queue,  and  you  walked  through  this  line  being  badgered 
all  the  while  by  the  ROTC  [Reserve  Officer  Training  Corps]  people, 
trying  to  make  sure  that  you  had  enlisted  in  the  ROTC.   And  when 
you  got  to  the  tally,  they  would  look  up  on  the  board,  and  if  the 
course  was  closed  that  was  it.   So  I  spent  two  days  of  never 
getting  a  schedule. 

Finally,  for  those  that  were  left  over,  you  took  whatever 
leavings  there  were.  And  so,  I  would  have  a  course  at  eight 
o'clock  in  the  morning,  and  another  one  at  one  o'clock  in  the 
afternoon.   Of  course,  coming  from  a  place  that  was  two  and  a  half 
hours  away,  an  eight-o'clock  class  meant  I  left  home  at  the  crack 
of  dawn.   So,  I  did  that  for  three  days,  and  1  decided  it  wouldn't 
work. 

The  first  class  1  went  to  was  a  physics  class.   Now,  I  had 
come  from  a  high  school  which  was  not  specially  oriented  towards 
science,  certainly  not  to  the  extent  of  several  special  high 
schools  in  Brooklyn  at  that  time,  one  of  them  called  Brooklyn 
Tech,  which  was  very  much  oriented  towards  people  going  into 
engineering  or  science.   As  high  school  students,  they  had  already 
had  many  of  the  kinds  of  courses  that  anybody  else  would  take  when 
they  went  to  a  university. 

In  the  first  physics  class  I  sat  in  on,  we  were  given  an 
exercise  to  make  some  measurements.   They  were  trying  to  teach  us 
something  about  the  variables  in  making  repetitive  measurements 
and  how  to  determine  the  significance  of  the  measurements.   There 
were  these  blocks  of  wood  on  the  table,  and  some  kind  of  a  tool 
that  I  had  never  seen  before,  and  a  data  pad.   I  was  told  to  start 
making  these  measurements,  and  I  didn't  even  know  what  this  tool 
was  or  how  to  use  it.   I  looked  over  at  people  sitting  on  either 
side  of  me,  and  they  were  going  ahead  and  measuring.   When  I  asked 
somebody  if  he  could  show  me  how  to  use  it,  I  got  brushed  off  very 
abruptly.   So  it  was  clear  that  the  atmosphere  there  was  very 
intense,  highly  competitive,  and  I  would  say  certainly  not 
friendly. 


Biology  Student  at  the  Brooklyn  College  Campus 


Berg:    I  didn't  like  the  prospect  of  continuing  that  for  I  don't  know  how 
long,  so  I  withdrew,  and  I  went  to  another  of  the  City  Colleges, 
which  is  the  Brooklyn  College  campus,  much  closer  to  home.   I 
enrolled  as  a  biology  major,  since  I  knew  chemical  engineering  was 


10 


definitely  not  what  I  wanted.   Especially  since  in  one  of  the 
lectures,  somebody  talked  about  what  chemical  engineers  do,  which 
was  building  factories  to  manufacture  various  kinds  of  chemicals, 
and  designing  the  machinery  for  carrying  out  certain  processes.   I 
realized  that  was  not  what  I  wanted.   I  actually  was  more 
interested  in  bench  research.   So,  I  decided  I  would  go  into 
biology. 

Biology  was  exceedingly  disappointing.   It  was  the  classical 
thing  where  you  dissect  some  little  pickled  guinea  pig  or 
something,  and  then  make  exotic  drawings  of  everything  in  it. 
Totally,  totally  boring.   And  so  there  I  was,  really  disappointed. 
I  didn't  like  the  chemistry;  I  didn't  like  the  biology—what  to 
do? 


Biochemistry  Student  at  Pennsylvania  State  University,  19A3-19A8 


Berg:    At  that  time  a  friend  of  mine,  who  also  lived  in  Seagate  and  was 
an  engineering  student,  went  off  to  Penn  State.   He  sent  me  a 
catalog  of  Penn  State,  and  I  thumbed  through  it,  and  lo  and 
behold,  there  was  something  called  biochemistry,  the  Department  of 
Biochemistry.   This  was  not  chemistry  and  it  wasn't  biology,  but 
it  looked  like  it  was  what  I  was  really  interested  in  the 
processes  that  go  on  in  living  organisms.   So,  1  decided  to  go  to 
Penn  State. 

Now  that  I  think  about  it,  I  think  I  graduated  in  January, 
1943,  and  attended  Brooklyn  College  in  the  spring  term.   That 
summer,  which  would  be  the  summer  of  1943,  I  turned  seventeen,  and 
I  was  allowed  to  enlist.   I  went  off  to  Penn  State,  waiting  for 

the  navy  to  call  me. 

Hughes:   Was  it  a  full  department  of  biochemistry? 

Berg:     Yes. 

Hughes:   That  was  a  bit  unusual. 

Berg:    It  was  unusual.   But  it  was  not  unusual,  because  it  was  in  the 
Agriculture  School,  which  is  where  biochemistry  was  largely 
centered  in  the  thirties  and  forties.   Most  of  the  focus  of 
biochemistry  at  that  time  was  on  analytical  chemistry  of 
biological  materials.   The  people  that  came  out  of  that  program 
generally  ended  up  in  the  pharmaceutical  industry  or  in  the  food 
industry.   In  fact,  while  I  was  going  to  college,  I  was  working 


11 


summers  in  food  companies,  in  their  research  labs,  doing  what  they 
call  today  food  technology. 

The  other  kind  of  biochemistry  was  largely  in  medical 
schools,  and  it  was  generally  called  physiological  chemistry.   But 
I  didn't  know  this  Department  of  Biochemistry  was  in  the  ag 
school.   It  didn't  make  any  difference  because  within  a  few  months 
I  was  called  up  in  the  navy.   So,  I  completed  one  semester  at  Penn 
State  as  a  freshman  student  majoring  in  biochemistry. 


Military  Service  in  World  War  II 


Berg:    It  was  the  navy  procedure  to  send  you  back  to  the  university  for 
further  training.   If  you  had  already  been  to  the  university,  you 
were  permitted  to  continue  in  whatever  you  had  been  studying,  and 
doing  in  addition  the  few  courses  that  the  navy  wanted  you  to 
take,  which  were  navigation  and  a  few  different  engineering 
courses.   If  you  had  never  been  to  the  university  before-- 


Berg:    --you  were  immediately  sent  to  a  place  and  put  into  an  engineering 
curriculum.   People  generally  stayed  in  these  places  for  about  a 
year,  before  they  were  to  go  on  to  the  next  stage  of  the  flight 
training  program. 

Hughes:   That  was  a  stroke  of  luck. 

Berg:    That  was  a  big  stroke  of  luck.   Originally,  I  was  sent  to 
Middlebury  College  in  Vermont.   But  I  knew  that  the  usual 
procedure  was  that  if  you  had  already  started  at  a  university,  and 
it  had  a  navy  unit,  you  were  usually  sent  to  the  place  where  you 
had  been.   So,  I  wrote  to  the  navy  and  asked  them  if  I  could  be 
reassigned  to  Penn  State,  which  I  was.   So  when  I  went  back,  I  was 
wearing  a  navy  uniform,  and  I  lived  in  a  navy  barracks  on  the 
campus.   All  the  courses  that  I  took  were  the  same  courses  I  would 
have  taken  had  I  been  a  civilian,  and  that  was  fortunate.   I  did 
that  for  three  semesters,  I  guess  it  was,  one  entire  calendar 
year. 

By  that  time  it  would  be  late  1944;  the  navy  decided  that 
the  attrition  of  navy  pilots  was  far  smaller  than  they  had 
anticipated,  and  so  many  of  us  were  transferred  from  the  navy 
flight  training  program  into  what  they  call  the  deck  officer 
school.   I  was  sent  to  a  midshipman  school  to  be  trained  for  ship 
duty.   And  that  was  in  New  York  City  out  on  Long  Island  Sound,  a 


12 


place  called  Fort  Schuyler.   We  were  what  people  came  to  call  the 
ninety-day  wonders.   You  went  for  three  months  and  you  were 
commissioned  as  an  ensign  in  the  navy. 

I  was  assigned  to  a  sub  chaser  program.   And  I  went  off  then 
to  this  place  in  Key  West  which  trained  officers  for  duty  on 
submarine  chasers.   Those  kinds  of  ships  that  I  was  on  did  either 
convoy  duty,  which  was  to  protect  maritime  shipping,  chasing 
submarines,  or  controlling  landing  ships  during  an  invasion.   So, 
I  didn't  get  to  do  what  I  wanted  to  do,  which  was  why  I  enlisted; 
I  wanted  to  fly,  but  I  ended  up  in  the  navy  doing  sea  duty.   I 
stayed  in  the  navy  until  after  Hiroshima.   Then  I  spent  one  year 
bringing  ships  back  from  the  Pacific  and  various  parts.   In  the 
summer  of  "46,  I  was  released  from  the  navy. 

Hughes:   You  were  in  the  Pacific? 

Berg:     Yes,  and  also  I  was  on  ships  that  were  protecting  convoys  in  the 

Caribbean.   I  don't  think  that  most  people  really  ever  appreciated 
how  much  American  shipping  was  lost  to  the  German  submarines  that 
were  literally  stationed  off  the  East  Coast  and  particularly  the 
Caribbean,  because  there  was  a  lot  of  traffic  through  the  Gulf  of 
Mexico.   They  devastated  American  shipping. 

Our  job  was  to  try  to  reduce  that  devastation.   By  that 
time,  of  course,  the  navy  had  already  broken  some  of  the  German 
naval  code.   They  knew  where  the  submarines  were,  and  so  it  wasn't 
quite  as  devastating.   Britain  came  very,  very  close  to  losing  the 
war  early  on,  because  supplies  to  them  almost  never  got  there,  it 
was  so  bad.   Until  they  broke  the  naval  code,  which  is  a  story  in 
itself,  they  were  on  the  verge  of  desperation.   But  once  they 
broke  the  code,  they  knew  where  the  German  submarines  were,  and 
they  were  able  to  avoid  them. 

When  the  war  ended,  there  were  a  lot  of  ships  that  had  to  be 
brought  back  from  the  Pacific.  And  so  we  brought  them  back,  and 
then  took  them  into  what  they  call  mothballing;  storing  them  in 
various  tributaries.   And  then  they  just  covered  them  over  with 
this  protective  covering  to  presumably  preserve  them.   I  was 
released,  as  I  say,  in  the  summer  of  "46,  and  went  back  to  Penn 
State  that  fall. 


13 


Return  to  Penn  State 


Marriage 


Berg:    Although  I  had  been  in  the  navy  for  close  to  three  years,  I  hadn't 
really  lost  that  much  time.   I  only  had  two  years  to  finish 
college.   And  I  did  that,  and  finished  with  a  major  in 
biochemistry.   I  got  married  somewhere  along  the  line,  actually  at 
the  end  of  the  junior  year  [1947],  to  somebody  I  had  known  for 
years  and  years,  although  she  was  not  from  where  I  grew  up. 
Millie  and  I  had  worked  together  one  summer  in  New  York  City  in  a 
company  in  which  her  father  was  one  of  the  executives,  and  1  was 
the  office  boy,  and  she  was  the  mail  clerk.   [laughter]   I  was 
sixteen  and  she  was  fifteen. 

Hughes:   That's  quite  a  story. 

Berg:     We  kept  in  touch  with  each  other  while  I  was  in  the  navy,  and  when 
I  came  back  to  New  York,  sometimes  we'd  get  together.   But  after  1 
got  out  of  the  navy,  we  established  contact  and  then  we  were 
married  the  next  year,  in  19A7.   So  we're  coming  up  to  our 
fiftieth  wedding  anniversary  this  fall. 


Decision  to  Do  Graduate  Work  in  Biochemistry 


Berg:     Those  two  years  at  Penn  State  were  rather  interesting,  because 

that's  when  I  really  started  doing  biochemistry  in  a  serious  way. 

Hughes:   Why? 

Berg:    First  of  all,  I  was  a  little  older  than  I  would  have  been  had  I 
just  gone  straight  through.   I  was  married  now,  and  really  much 
more  focused  on  getting  out  of  school.   I  knew  I  wanted  to  go  on 
to  graduate  school.   I  didn't  know  where  or  how.   I  had  decided 
that  medicine  was  not  what  I  wanted  to  do.   I  did  want  to  do 
research. 

At  the  end  of  my  junior  year,  I  worked  for  General  Foods 
Corporation,  over  in  Hoboken,  New  Jersey,  in  their  research  labs, 
doing  just  standard  analytical  work.   The  following  year,  I  worked 
at  the  Lipton  Tea  Company  in  their  research  laboratories,  also  in 
Hoboken,  again  doing  largely  food  technology. 


14 


Hughes:   What  effect  did  those  summer  jobs  have? 

Berg:    Well,  the  summer  jobs  had  one  strong  motivating  force.   You  could 
see  that  the  people  who  had  bachelors  degrees  were  the  ones  who 
were  being  told  what  to  do.  And  the  people  who  were  telling  them 
what  to  do  were  people  with  Ph.D.s.   Very  quickly  that  was  the 
deciding  point,  that  if  1  was  going  to  go  into  science,  I  did  not 
want  to  be  in  a  position  where  people  were  telling  me  what  to  do. 
I  wanted  to  be  able  to  drive  the  research  myself.   And  that's  when 
I  decided  that  I  wanted  to  do  a  Ph.D.   So  it  was  the  end  of  the 
junior  year,  when  I  knew  I  was  going  to  go  on  to  graduate  school. 

My  wife  was  a  registered  nurse.   In  fact,  during  the  war  she 
had  been  in  nurses  training.   And  when  she  graduated  in  1947,  we 
were  married.   She  went  back  to  State  College,  which  is  where  Penn 
State  is.   State  College  was  a  town  of  about  three  thousand 
people,  a  dinky  little  place—no  hospital.   We  were  living  in  a 
rented  room  in  somebody's  rooming  house.   She  tried  to  find  a  job, 
and  a  hospital  was  twelve  miles  away.   We  had  no  car.   She 
obviously  couldn't  do  that.   So  she  worked  for  a  while  looking 
after  newborn  babies  born  to  student  wives  during  the  first  weeks 
after  birth.   That  was  clearly  not  satisfying.   So  she  went  back 
to  New  York  City,  lived  with  my  parents,  and  worked  in  a  local 
hospital  there.   So  for  that  last  year,  we  were  separated.   1  used 
to  commute  back  and  forth  from  State  College,  about  an  eight-hour 
train  trip,  to  New  York  for  the  weekends  and  then  go  back  on 
Sunday  nights.   And  that  was  a  hassle. 

During  my  last  year,  again,  my  performance  in  school  was 
pretty  good.   I  was  identified  pretty  quickly  as  a  bright  young 
student  to  be  helped.   I  wrote  several  papers,  not  for 
publication,  but  during  my  last  year,  1948.   One  of  them  was  about 
the  newly  emerging  use  of  isotopes  for  tracing  metabolic 
reactions,  and  that  excited  the  heck  out  of  me.   I  gave  several 
talks  on  it. 


Graduate  Student.  Western  Reserve  University,  1948-1952 
Applying  to  Graduate  Schools 

Berg:    So  I  applied  to  this  one  school,  which  seemed  to  be  the  place  from 
which  many  of  the  review  articles  and  published  papers  were 
coming.   It  was  Western  Reserve  University,  a  place  I  had  never 
heard  of.   I  thought  it  was  an  Indian  reservation.   I  wrote  to 


15 

them,  and  they  didn't  have  an  opening  for  a  new  research 
assistant,  a  graduate  student. 

Hughes:   Who  was  there? 

Berg:    A  man  named  Harland  Wood. 

Hughes:   He  was  the  one  using  the  isotopes? 

Berg:    He  had  just  created  a  new  department.   I'll  come  back  to  that  in  a 
little  bit,  because  it's  a  very  important  part  of  my  life. 

During  that  last  year,  knowing  that  I  wanted  to  go  to 
graduate  school,  I  approached  it  in  my  typical  way,  which  is,  I 
got  a  book  about  all  the  graduate  schools  that  had  biochemistry 
departments,  and  I  wrote  a  letter  to  each  one  of  them  applying  for 
admission.   I  must  have  sent  out  about  sixty  letters. 

It  was  the  same  way  I  got  the  summer  jobs.   I  would  write  to 
every  chemical,  biology,  drug  company,  food  company  in  the  New 
York  Metropolitan  area,  and  just  wait  for  an  answer.   [laughter] 
And  I  always  got  an  answer  offering  me  a  job  for  the  summer.   And 
so  I  did  the  same  thing  for  graduate  school,  just  applying  to  a 
large  number  of  places.   Not  unexpectedly,  some  of  them  had 
biochemistry  departments  in  the  medical  school,  others  in  the 
agriculture  school. 

So,  I  applied  to  Western  Reserve;  it  was  one  of  the  many 
that  I  applied  to.   It's  an  interesting  fact  relating  to 
Kornberg's  talk  about  anti-Semitism:  I'd  get  back  some  letters 
which  were  offering  me  admission  into  the  graduate  program  in 
biochemistry,  with  a  warning  or  statement  telling  me  that  by  no 
means  should  I  consider  this  as  a  possibility  that  I  would  be  able 
to  enter  the  medical  school  through  the  back  door.   They  were 
really  quite  up  front  about  that. 

Eventually,  I  chose  to  go  to  a  place  called  Oklahoma  A  and 
M,  which  today  is  called  Oklahoma  State  University.   For  a  guy 
from  Brooklyn  to  choose  to  go  to  Oklahoma  A  and  M  was  already 
pretty  radical.   [laughter]   But  part  of  the  reason  was  the 
department  of  chemistry  offered  us  a  place  to  live,  found  a  job 
for  my  wife  to  work  in  the  local  hospital,  and  they  were  really 
recruiting  heavily.   So  I  agreed  to  go. 

Hughes:   You  went  for  those  somewhat  peripheral  reasons,  not  for  the 
science? 

Berg:    That's  right.   It  was  known  as  a  pretty  good  school  of  chemistry, 
but  I  didn't  know  a  lot  more  about  it.   And  so  I  accepted. 


16 


Decision  to  Attend  Western  Reserve  University 


Berg:    Within  about  two  weeks  of  the  time  we  were  going  to  leave  Penn 
State,  and  planning  to  go  west,  I  got  a  call  or  letter  from 
Western  Reserve  saying  that  they  now  had  an  opening,  and  they  had 
money,  and  if  I  would  like  to  come,  it  would  be  worked  out.   I  was 
very,  very  troubled  because  I  had  already  accepted  this  very 
generous  offer  from  Oklahoma  A  and  M,  and  I  remember  being  very 
troubled  about  turning  it  down. 

I  went  to  see  both  the  dean  and  the  chairman  of  the 
biochemistry  department  at  Penn  State,  and  to  this  day,  I  can 
never  thank  them  enough  for  having  said,  "Look,  just  call  up 
Oklahoma  A  and  M  and  tell  them  straight  off  that  you've  had  your 
mind  set  on  doing  this  kind  of  work,  and  here  was  an  opportunity 
to  do  that."   I  did  that  and  they  were  extraordinarily  gracious. 
I  was  so  bowled  over,  because  it  was  so  unexpected.   I  thought 
they  would  give  me  a  hard  time  by  saying  how  much  difficulty  they 
had  gone  through  to  get  me  settled. 


The  Department  of  Clinical  Biochemistry 


Berg:     In  any  case,  when  I  got  to  Western  Reserve  in  Cleveland  in  1948, 
what  I  found  is  that  I  had  been  accepted  by  the  wrong  department, 
[laughter]   In  fact,  what  I  had  actually  applied  to  was  an  ad  in 
the  Chemical  and  Engineering  News,  in  the  back  section  where  they 
printed  job  openings.   And  it  said,  "Western  Reserve  University, 
Department  of  Clinical  Biochemistry,  Medical  School."  And  since  I 
recognized  that  Western  Reserve  University  was  the  place  from 
which  all  these  papers  were  coming  that  I  was  so  excited  about,  I 
just  assumed  that  they  were  one  and  the  same. 

The  biochemistry  department  at  Western  Reserve  University 
had  been  headed  by  a  man,  Victor  Meyers,  since  1915  or  "20,  who  by 
1948  was  literally  senile  and  long  since  beyond  any  period  when  he 
had  been  productive.  When  the  war  ended,  Western  Reserve 
University  realized  they  had  to  rejuvenate  the  biochemistry 
department .   So  they  recruited  from  Iowa  what  was  then  one  of  the 
hottest  young  biochemists  in  the  country,  named  Harland  Wood.   He 
brought  his  entire  group  from  Iowa,  and  several  people  who  were  in 
Minnesota.   He  created  a  department  essentially  in  one  fell  swoop. 
What  to  do  with  the  man  who  had  been  head  of  the  department?  They 
created  Clinical  Biochemistry,  of  which  Victor  Meyers  was  the  sole 
occupant. 


17 


So,  when  I  got  there,  I  was  told  I  was  going  to  be  in  this 
department,  and  I  realized  that  it  had  nothing  to  do  with  the  one 
I  wanted,  which  was  on  the  floor  above.   But  there  wasn't  much  I 
could  do;  I  was  there.  And  when  I  went  to  see  Victor  Meyers  to 
discuss  what  I  was  going  to  do,  he  gave  me  a  couple  of  old  theses, 
which  were  by  people  who  had  analyzed  cholesterol  levels  in 
various  tissues  of  autopsied  individuals.  And  my  assignment  was 
to  do  the  cholesterol  level  in  some  eighty-five  postmortem  heart 
muscles  and  see  if  I  could  correlate  cholesterol  level  with  cause 
of  death.   Well,  that  was  pretty  discouraging. 


Research  on  the  Artificial  Kidney 


Berg:    Fortunately,  Meyers  died  about  a  month  later,  and  so  there  I  was 
stuck  in  this  pseudo-department.   But  two  of  Meyers'  earlier 
students  were  working  in  the  lab,  and  one  of  them,  Jack  Leonards, 
had  been  given  a  junior  appointment  in  the  department  of 
biochemistry,  largely  as  a  courtesy.   He  asked  me  if  I  would  be 
willing  to  stay  on  and  work  with  him.   Well,  I  didn't  have  much 
choice,  so  I  said  yes. 

It  turned  out  that  he  and  this  other  person,  Leonard  Skaggs, 
who  had  also  been  a  former  student  there,  were  developing  an 
artificial  kidney.   At  that  time  there  was  already  a  precursor, 
which  had  been  developed  by  a  Dutchman  named  Kolff,  and  which  was 
in  use,  but  was  very,  very  complicated.   It  was  a  big  rotating 
drum  around  which  dialyzing  tubing  was  wrapped,  and  blood  ran 
through  the  tubing,  and  this  drum  rotated  it  through  a  bath  which 
allowed  dialysis  to  occur.   So  toxic  substances  that  were  in  the 
blood  could  pass  through  the  membrane  into  this  bath. 

These  two  guys  had  come  up  with  a  design  which  was  sort  of  a 
neat  little  box.  It  was  a  very  ingenious  idea.  And  so  I  said,  I 
have  nothing  to  lose;  I'd  work  for  them.  So  I  started  to  use  the 
artificial  kidney  to  learn  how  to  keep  animals  alive  after  you've 
taken  their  kidneys  out.  I  had  to  learn  a  lot  of  surgery  and 
things  I  wasn't  terribly  interested  in. 


Joining  the  Department  of  Biochemistry 


Berg:    Meanwhile,  I  was  taking  courses  that  graduate  students  had  to 

take.   All  the  courses  were  in  the  biochemistry  department,  plus 
the  first  two  years  of  medical  school  courses.   Well,  one  of  the 


18 


courses  I  had  to  take  up  in  the  biochemistry  department  was  a 
course  in  which  students  were  asked  to  give  presentations  on 
current  research  papers,  and  I  gave  one  or  two  of  them.   And  not 
long  afterwards,  I  was  approached  by  the  chairman  of  the 
department:   Would  I  consider  moving  up  into  the  biochemistry 
department?  Well,  that  was  the  kind  of  thing  you'd  say,  "I 
thought  you'd  never  ask." 

So,  at  that  point,  I  went  to  these  two  fellows  and  told  them 
that  I  had  always  had  my  heart  set  on  working  in  this  lab  where 
there  was  use  of  isotopes,  and  not  on  this  type  of  physiological 
chemistry  that  they  were  doing.   They  were  very  good  about  saying, 
"By  all  means,  go."  And  so  I  moved  up  to  the  department  and 
became  a  graduate  student  there. 

I  had  already  spent  two  years  in  this  process  and  at  that 
time  a  Ph.D.  program  was  normally  four  years.   People  did  not  take 
longer.   And  so  I  essentially  did  my  thesis  research  in  two  years. 
Our  students  today  take  five,  six  years  and  don't  do  half  what  we 
did,  because  we  had  to  teach,  and  we  had  to  run  the  laboratories 
for  the  medical  students  and  take  a  lot  of  courses.   We  had  to 
take  almost  the  entire  curriculum  for  the  first  two  years  of 
medical  school,  even  though  we  were  not  going  to  go  on  and  be 
doctors . 

Harland  Wood  and  the  people  in  the  department  were  different 
role  models  than  anything  I  had  seen.   First  of  all,  it  was  an 
academic  setting.   It  wasn't  an  industrial  lab.   And  the  pace  of 
discovery  in  my  research  went  extremely  well.   I  received  a  lot  of 
kudos  and  gave  talks  at  national  meetings,  and  so  on.   The 
feedback  I  got  was  that  I  could  do  this;  this  is  a  career  that  I 
could  be  successful  in.   And  it  looked  like  it  was  great  fun,  to 
just  go  into  a  lab  and  be  able  to  do  whatever  you  wanted. 
Teaching  was  not  a  difficult  thing,  and  jobs  were  plentiful. 

Hughes:   So,  you  were  teaching  while  you  were  learning. 

Berg:     That's  right. 

Hughes:   You  hadn't  had  much  formal  biochemistry,  had  you? 

Berg:    Well,  what  we  did  was  teach  in  the  laboratories.   Biochemistry 

courses  had  laboratories  associated  with  them.   Medical  students 
had  to  go  in  and  carry  out  various  exercises.   There  were  maybe 
twenty  or  thirty  experiments  over  a  term,  and  you  were  there  to 
help  them.   We  had  to  set  up  the  reagents  for  them.   We  had  to 
test  the  experiments  to  see  if  they  would  work,  and  things  of  that 
sort.   And  we  had  to  spend  long  hours  in  the  laboratory  with  the 
students.   And  then  when  the  laboratory  was  over,  we  could  go  and 


19 


work.   The  general  pattern  of  working  for  a  graduate  student  is 
essentially  into  the  wee  hours  of  the  morning. 

My  wife  was  a  nurse  at  Western  Reserve  Medical  Center,  a  big 
hospital  which  was  part  of  the  medical  center.   And  she  chose  to 
work  some  very  interesting  hours,  like  six  to  midnight.   That 
meant  she  had  the  day  free,  and  I  could  go  back  in  the  evening  and 
work  until  midnight.  And  then  I'd  go  pick  her  up  from  the 
hospital,  and  then  we'd  go  do  something  sometimes.   That  was  a 
very  good  thing,  because  the  hospital  had  a  hard  time  getting 
people  to  work  those  hours,  so  she  only  had  to  work  six  hours  and 
was  paid  for  eight,  which  was  nice.   She  largely  supported  the  two 
of  us.   I  was  on  the  G.I.  Bill,  which  I  think  was  something  like 
$165  a  month.   I  don't  know,  her  salary  was  maybe  twice  that.   And 
so  we  lived  on  that. 

We  had  an  apartment  very  near  the  medical  center,  so  we 
could  both  walk  to  the  lab  or  to  the  hospital.   We  spent  four 
years  in  Cleveland,  which  actually  was  great  fun.   We  really 
enjoyed  it,  made  a  lot  of  friends.   The  principal  thing  about  it 
was,  it  opened  up  new  vistas  for  what  I  wanted  to  do. 


Research  on  Nutritional  Supplements 


Hughes:   What  was  the  research? 

Berg:     It  was  to  solve  a  problem  that  had  really  been  a  central  theme  or 
problem  in  biochemistry.   Animals  on  certain  diets,  for  reasons 
that  nobody  could  tell,  would  die.   Well,  that's  not  a  very  good 
explanation.   There  were  certain  compounds  or  nutrients  that  were 
thought  to  be  an  essential  part  of  every  person's  diet.   And  if 
you  left  them  out  of  the  diet,  they  led  to  death.   One  was  the 
amino  acid,  methionine,  and  a  compound  called  choline.   If 
methionine  was  omitted  from  the  diet,  animals  usually  developed 
very  fatty  livers  and  then  ultimately  died. 

About  the  time  that  I  was  plugging  away  at  dialyzing 
animals,  one  of  the  great  figures  in  biochemistry,  a  man  by  the 
name  of  Vincent  du  Vigneaud,  was  a  professor  of  biochemistry  at 
Cornell  Medical  School,  and  later  received  the  Nobel  Prize  [1953]. 
He  was  one  of  the  ones  that  had  reported  that  methionine  was  an 
essential  part  of  the  diet.   But  there  were  hints  that  this  wasn't 
necessarily  always  true,  because  if  you  varied  the  diet, 
supplemented  it  with  certain  vitamins,  you  could  leave  methionine 
out,  and  the  animals  did  fine.   The  two  things  were  folic  acid  and 
vitamin  B-12.   When  you  looked  in  the  tissue  in  the  diet,  what  you 


20 


found  is  that  the  animals  contained  methionine  and  choline  which 
previously  had  to  be  supplied  in  the  diet.   So,  it  was  obvious 
that  given  these  vitamin  supplements,  they  were  able  to  completely 
synthesize  these  molecules,  which  had  been  thought  to  be  unable  to 
be  synthesized. 

There  were  already  some  indications  of  what  kind  of 
molecules  could  be  used  as  the  building  blocks  to  build  methionine 
and  choline.   One  of  the  people,  Warwick  Sakami,  in  the 
biochemistry  department  where  I  was  had  done  an  experiment 
injecting  radioactive  formaldehyde  into  rats  and  then  recovering 
methionine  and  choline  from  their  livers.   He  showed,  using  this 
radioactive  formaldehyde,  that  the  methyl  groups  of  methionine  and 
choline  had  been  produced  using  the  formaldehyde. 

One  of  the  great,  great  things  about  using  radioisotope 
tracers  was  that  you  could  feed  a  molecule  that  was  marked 
radioactive,  and  you  could  determine  whatever  it  ended  up  in,  the 
new  molecule  because  it  contained  radioactivity.   So,  formaldehyde 
had  been  converted  into  the  methyl  groups  of  methionine  and 
choline.   And  so,  it  looked  like  these  animals,  when  provided  with 
the  appropriate  vitamins,  were  actually  able  to  convert  the  one- 
carbon  compound,  formaldehyde,  and  subsequently  formic  acid  and 
methyl  alcohol.   All  three  of  these,  in  fact,  could  eventually  be 
converted  in  the  body  to  the  methyl  groups  of  methionine  and 
choline . 


Berg's  Initial  Research  Project 


Berg:    In  one  of  the  seminars,  when  I  was  still  a  student  in  Clinical 
Biochemistry,  1  reported  this  progress.  And  in  preparing  the 
seminars,  it  came  to  me  that  there  were  certain  kinds  of 
experiments  that  could  be  done  that  would  test  how  this  synthesis 
went.   And  so,  I  went  to  Warwick  Sakami,  in  biochemistry,  and  I 
told  him  that  it  seemed  to  me  that  one  could  find  out  something 
about  this  process,  by  the  following  kind  of  experiment.   And 
that's  when  he  said,  "How  would  you  like  to  come  up  here  to  the 
biochemistry  department  and  do  it?"  And  then  he  went  to  the 
chairman  of  the  department,  and  Harland  Wood  then  asked  me  if  I 
would  be  interested  in  coming  up.   So,  when  I  went  up,  I  did  this 
experiment,  and  it  came  out  beautifully,  describing  for  the  first 
time  how  this  one-carbon  molecule  could  get  into  these  other 
products . 

Hughes:   So  they  were  intrigued  by  the  science  that  you  proposed  to  do. 
But  were  they  also  impressed  by  the  fact  that  you  were  using  a 


21 

technique  that  Harland  Wood  was  interested  in,  namely  the  use  of 
radioisotopes? 

Berg:    Everybody  in  the  department  was  doing  that. 
Hughes:   Using  radioisotopes. 

Berg:    Using  radioisotopes.   So,  I  didn't  bring  anything  novel  to  the 
idea  of  using  radioisotopes.   It  was  more  how  to  use  the 
technology  to  actually  explain  an  interesting  and  at  that  time 
unknown  biological  process.   So  my  recollection  of  what  they  said 
is,  "That's  a  neat  idea.   Why  don't  you  do  it?"  So,  I  went  up  and 
set  up  the  experiments  and  actually  started  to  do  them.   And  as  I 
say,  they  came  out  pretty  much  the  way  I  predicted. 

That  then  led  me  to  try  and  do  enzyme  experiments,  asking 
could  we  demonstrate  the  enzymes  that  carry  out  this  process? 
Well,  at  that  time,  I  wasn't  very  much  of  an  enzymologist .   All  I 
could  do  was  set  up  these  tissue  extracts.   They  were  relatively 
crude  extracts.   But  we  showed  we  could  convert  formic  acid  or 
formaldehyde  to  the  methyl  groups  of  methionine  and  choline  in  a 
cell-free  system. 

Hughes:   Was  that  something  that  people  with  a  biochemical  background  would 
have  known  how  to  do? 

Berg:     You  mean  how  to  do  the  extracts? 
Hughes:   Yes. 

Berg:    Yes,  doing  that  was  not  so  novel.   Kornberg  was  doing  enzymology 
long  before  that.   People  were  studying  how  one-carbon  compounds 
are  used  to  make  purines  in  nucleic  acids.   So,  what  I  did  was 
actually  set  up  for  the  first  time  an  in  vitro  system  that  could 
actually  manufacture  methionine  and  choline,  and  I  used  these 
radioactively  tagged  molecules  to  show  how  that  conversion  went. 
I  was  able  to  show  that  in  fact  in  these  extracts  there  was 
evidence  that  you  needed  cof actors,  so-called,  which  were  derived 
from  folic  acid,  which  had  been  hinted  at  by  showing  that  animals 
fed  high  levels  of  folic  acids  and  B-12  could  make  everything. 
So,  it  was  not  terribly  surprising.   I  published  several  papers  on 
that.1 


1  For  references  throughout  the  oral  history  to  Dr.  Berg's 
publications,  see  his  bibliography  in  the  appendix. 


22 


Countering  du  Vigneaud  iti 


Berg:    One  of  the  things  that  happened  was,  I  went  to  a  national  meeting, 
and  when  I  presented  this  work,  Professor  du  Vigneaud,  this  man 
who  had  been  the  real  honcho  for  this  whole  field,  got  up  and  gave 
me  a  hard  time.   He  had  been  protecting  the  principle  and  concept 
that  these  substances  were  required  in  the  diet.   And  here  was 
somebody  getting  up  and  showing  that  they  were  not  only  not 
required,  but  here  was  a  way  in  which  they  could  be  made  in  the 
body. 

I  must  have  handled  myself  very  well  in  this  debate,  because 
plenty  of  people  told  me  about  it  afterwards.   But  interestingly 
enough,  du  Vigneaud  went  to  Harland  Wood  and  said,  "You  have  a 
young  professor  in  your  department,  Paul  Berg.   I  want  to  offer 
him  a  job  here  at  Cornell  in  New  York."  And  Wood  told  him,  "He's 
not  a  professor;  he's  a  graduate  student."   [laughter]   So,  my 
work  really  flourished;  it  went  very  well.   And  by  the  time  I  was 
ready  to  go,  I  was  already  known  somewhat  outside  Western  Reserve. 


Harland  Wood  and  Radioisotopic  Tracers 


Berg:    Now,  there  was  a  steady  parade  of  distinguished  people  who  came 
through  Western  Reserve  because  Harland  Wood  was  one  of  the  very 
leading  top  people  in  this  field.   He  had  developed  whole  new 
approaches  to  studying  metabolism  using  these  isotopes.   And  he 
had  invented  instruments  that  made  it  possible  to  study  both 
radioactive  and  stable  isotopes  —  stable  isotopes  like  carbon-13. 
He  had  built  a  mass  spectrometer  to  do  this  kind  of  analysis.   He 
was  a  remarkable  guy. 

Hughes:   Postwar,  radioisotopes  were  relatively  easy  to  obtain? 

Berg:     Easy  to  obtain  because  Oak  Ridge  Laboratory  was  cranking  them  out, 
and  they  were  commercially  available.   Radioactive  isotopes 
carbon-14  and  P  [phosphorus] -32  were  available.   The  other 
isotopes  were  much  harder  to  get,  but  they  too  could  be  obtained, 
because  they  were  made  by  some  form  of  enrichment.   Carbon-13  is 
normally  present  in  carbon  dioxide.   But  it's  present  in  only 
small  quantities,  and  nitrogen- 15  is  present  in  all  kinds  of 
nitrogen-containing  compounds,  again,  in  small  quantities.   During 
the  war  they  learned  how  to  enrich  for  these  heavy  isotopes,  as 
they  were  called.   And  the  radioactive  isotopes  were  made  in  a 
radiation  lab. 


23 

During  the  war,  before  radioisotopes  were  available,  Harland 
Wood  was  restricted  to  using  carbon- 13,  and  he  did  one  of  the 
great,  monumental  pieces  of  work  in  discovering  that  carbon 
dioxide  is  actually  metabolized.  We  build  lots  of  our  organic 
molecules  from  carbon  dioxide  that  we  had  always  considered  as  a 
waste  product. 

If  you  label  the  carbon  dioxide  and  feed  it  to  animals,  you 
can  show  that  they  in  fact  make  many  complex  molecules  from  the 
carbon  dioxide.   But  he  didn't  have  Geiger  counters  to  measure 
because  this  was  a  stable  isotope.   He  had  to  develop  a  mass 
spectrometer  that  measures  the  mass  of  atoms,  so  you  could 
distinguish  carbon- 13  from  carbon- 12,  which  are  the  natural  carbon 
isotopes . 

Wood  was  one  of  these  people  raised  on  a  farm  for  whom 
building  anything  was  not  an  impediment  to  doing  what  you  wanted 
to  do.   He  built  anything.   He  synthesized  all  his  radioactive 
compounds.   He  was  a  really  remarkable  guy,  and  he  was  a  wonderful 
person  so  that  you  were  strongly  attracted  to  him.   What  he 
represented  was  an  idea  of  what  you  would  love  to  be  like.   That's 
why  most  of  the  students  adored  him.   In  fact,  I  have  a  visit  next 
week  from  a  man  who  is  writing  his  biography. 

We  remained  very  close  friends  for  many,  many  years.   He 
died  about  five,  eight  years  ago.   But  he  continued  working  into 
his  eighties.   In  the  last  decade  of  his  life,  he  probably 
published  more  papers  than  the  rest  of  the  department  together. 
It  was  amazing.   We  went  to  his  seventieth  birthday  and  his 
eightieth  birthday. 

Hughes:   You  were  using  radioisotopes  mainly,  rather  than  stable  isotopes? 

Berg:     Yes. 

Hughes:   And  getting  them  from  Oak  Ridge? 

Berg:     Yes,  you  could  buy  them.   You  bought  barium  carbonate,  which  was 
an  insoluble  compound.   It's  essentially  a  barium  salt  of 
carbonate,  BaC03.   Organic  chemists  had  worked  out  techniques  that 
converted  barium  carbonate  to  sodium  bicarbonate,  and  then  doing 
organic  chemistry  to  convert  the  sodium  bicarbonate  into  organic 
molecules.   And  we  had  to  do  all  that. 

Today,  students  just  go  to  the  catalogs  and  buy  everything; 
they  make  nothing.   But  we  had  to  synthesize  everything  that  we 
made.   And  so  a  lot  of  times  we  had  to  learn  the  chemistry  to  make 
methyl  alcohol,  how  do  you  make  propionic  acid,  or  pyruvic  acid, 
or  any  of  these  molecules  with  a  radioactive  carbon  atom  in  a 


particular  location  in  the  molecule?  These  were  fairly 
complicated  steps. 

Hughes:   Was  that  expertise  in  the  department? 

Berg:    Yes,  that  was  expertise  that  was  largely  in  the  department,  and  we 
got  lots  of  help.   Sometimes  you  had  to  work  out  a  procedure  that 
had  not  been  done  before.   So  you  made  your  precursor  molecules. 
You  either  fed  them  to  animals  or  injected  them  into  animals  or 
incubated  them  with  extracts.   And  then  you  recovered  the 
products,  and  then  you  had  to  determine  if  the  product  was 
radioactive.   That  was  easy.  And  then  you  had  to  ask  where  did 
this  radioactive  atom  end  up  in  the  molecule?   So  we  had  to 
develop  procedures  for  degrading  molecules  and  recovering  the 
different  pieces  in  a  form  where  you  could  measure  their 
radioactivity.   And  then  you  could  draw  the  chemical  reactions 
that  must  have  intervened  to  convert  this  into  that,  if  this 
molecule  ended  up  in  that  particular  part  of  the  other  molecule. 
And  so  we  had  to  develop  procedures  for  degrading  molecules  and 
recovering  the  different  pieces  in  a  form  where  you  could  measure 
their  radioactivity. 

Radioactivity  made  this  kind  of  experimentation  much  simpler 
than  using  stable  isotopes.   With  stable  isotopes,  you  had  to  have 
this  very  elaborate  mass  spectrometer,  and  you  had  to  recover 
larger  amounts  of  material.   Whereas  with  radioactivity,  you  could 
use  literally  trace  quantities. 

I  remember  very  clearly  once  having  to  make  methyl  alcohol 
from  barium  carbonate.   I  did  this  in  a  very  large  hood,  with  ten 
millicuries  of  radioactivity,  which  was  a  very,  very  large  amount 
of  radioactivity  at  that  time.   Today,  people  wouldn't  think  it 
was  very  much.   I  set  up  this  whole  organic  synthesis  array,  in 
which  the  starting  material  was  here,  and  the  products  had  to  go 
through  various  kinds  of  bubblers  and  be  collected  in  a  reservoir. 
I  had  to  build  the  glassware  myself.   And  when  I  did  the 
experiment,  and  I  looked  at  the  end,  I  had  nothing.   It  turned  out 
that  one  of  the  glass  bubblers  had  a  pinhole  in  it,  and  all  of  the 
gas  had  gone  through  the  hole.   I  had  lost  ten  millicuries  of 
carbon- 14,  for  which  I  was  ridiculed  and  got  a  lot  of  flack.   But 
that  was  the  trial  and  tribulation  of  learning.   [laughter] 


Visits  by  Arthur  Kornberg  and  Herman  Kalckar 


Berg:     I  remember  very  clearly,  Arthur  Kornberg  came  to  visit  Western 

Reserve  University  and  gave  a  lecture.   It  was  not  long  after  he 


25 

had  won  the  Paul-Lewis  Award  [1951]  for  his  work  in  enzymes.   This 
would  be  about  1951  or  '52.   His  picture  had  been  on  the  cover  of 
Chemical  and  Engineering  News.  At  that  time,  even  small  prizes 
got  you  a  lot  of  notoriety.   Today,  you  win  a  prize,  you  might 
earn  a  little  paragraph  in  the  back  of  the  journal. 

But  anyway,  when  I  read  about  Arthur,  I  discovered  that  he 
was  from  Abraham  Lincoln  High  School,  grew  up  in  Brooklyn,  and  so 
on.   And  so  after  he  gave  his  lecture,  Harland  Wood  had  a  cocktail 
party  at  his  home  for  him,  and  I  was  invited,  and  he  and  I 
chatted.   We  discovered  that  we  were  both  from  the  same  high 
school  and  the  same  background.   I  asked  him  if  there  was  a  chance 
that  I  could  come  work  in  his  lab  as  a  postdoc. 

Hughes:   You  knew  what  research  he  was  doing  because  you  had  gone  to  the 
lecture? 

Berg:    Oh  yes,  not  only  his  lecture  but  during  graduate  training  we 
studied  every  new  paper  that  came  out,  and  so  I  knew  in  great 
detail  what  he  had  been  doing.   It  was  the  kind  of  enzymology 
which  I  had  not  had  experience  with,  nor  was  there  anybody  in  the 
department  at  the  time  who  did  that  kind  of  enzymology. 

Hughes:   Which  was  what? 

Berg:    Well,  f ractionation,  obtaining  pure  enzymes.  We  were  content  to 

work  with  relatively  crude  preparations.   Kornberg  would  turn  over 
in  his  grave  if  he  knew  any  of  his  people  ever  worked  with  crude 
extracts. 

Hughes:   Was  he  fairly  unique  at  that  time  in  insisting  on  working  with 
pure  extracts? 

Berg:    Well,  I  think  he  was  one  of  a  small  number.   There  were  perhaps 
half  a  dozen  people  who  were  really  committed,  and  one  of  them 
made  the  point,  "Don't  waste  clean  thoughts  on  dirty  enzymes." 
Kornberg  had  taken  a  sabbatical  and  gone  to  work  with  a  man  named 
Carl  Cori,  who  was  one  of  the  giants  of  biochemistry. 

Berg:    There  were  lots  of  other  visitors,  and  one  of  them  was  a  man  who 

was  from  Denmark;  his  name  was  Herman  Kalckar.   Herman  Kalckar  was 
a  brilliant  scientist  and  had  made  major  discoveries  during  the 
late  thirties,  when  he  was  a  student  in  Copenhagen.   And  then  he 
came  to  this  country  on  a  Rockefeller  fellowship.   He  went  to 
Caltech,  and  then  the  war  broke  out  in  1939,  and  he  was  stuck  in 
the  States.   He  remained  at  Caltech  for  a  short  time  and  wrote  an 
extremely  influential  review  article,  which  gave  him  a  lot  of 
notoriety.   He  then  took  a  position  at  the  Public  Health  Research 
Institute  in  New  York,  and  he  spent  the  war  years  there.   He 


26 


developed  a  whole  new  approach  to  being  able  to  use  enzymes  in  a 
novel  way. 

Herman  came  and  gave  a  seminar  on  that  topic.   I  don't  know 
how  many  Danish  people  you  know,  but  many  of  them  are 
unintelligible.   He  was  more  unintelligible  than  most.   Their 
language  requires  a  lot  of  guttural  sounds  and  throat  sounds. 
When  Herman  spoke  English,  he  was  very  hard  to  understand,  and  he 
had  a  flamboyant  way  of  doing  it  that  made  it  even  more  difficult. 
So,  nobody  in  the  audience  knew  what  he  was  talking  about.   But  I 
loved  him;  I  thought  he  was  great. 

Hughes:   Did  you  understand  what  he  was  saying? 

Berg:    I  could  see  the  outlines  of  what  he  had  done,  and  it  was 

fascinating,  because  it  looked  like  it  was  a  new  way  of  using 
enzymology.   What  he  was  really  developing  was  an  analytical  way 
to  follow  enzyme  reactions. 

Hughes:   But  not  with  radioisotopes? 

Berg:     Not  with  isotopes.   This  was  using  a  spectrophotometer.   You  get 
spectral  changes  in  certain  molecules  when  the  enzyme  acts  on 
them,  and  therefore  you  can  follow  the  reaction  by  following  the 
spectral  changes.   At  that  time  there  was  a  very  new  instrument 
that  had  come  out  called  the  Beckman  spectrophotometer.   And  the 
Beckman  spectrophotometer  was  one  of  the  special  instruments  that 
everybody  sort  of  bowed  around.   And  here  Kalckar  was  using  this 
thing  in  this  very  clever  way,  and  it  just  struck  me.   Besides,  he 
seemed  like  an  interesting  personality.   When  the  war  ended,  he  of 
course  went  back  to  Copenhagen.   So,  I  talked  to  him  about  the 
possibility  of  going  to  work  in  his  lab,  and  he  said  that  was 
fine. 


Postdoctoral  Positions 


Deciding  Not  to  Go  to  the  Coris '  Lab 


Berg:    Meanwhile,  Harland  Wood  had  other  plans  for  me.   He  had  seen  me  as 
one  of  his  star  students,  and  he  felt  that  it  was  appropriate  for 
me  to  go  on  to  Carl  Cori's  lab,  and  that  it  was  a  fitting  place 
for  a  bright  young  person  to  go  and  get  the  next  level  of  his 
training.   Carl  Cori  was  at  Washington  University  in  St.  Louis, 
and  the  stories  I  heard  about  St.  Louis  made  it  sound  like  it  was 
unlivable . 


27 


Hughes:   The  city  itself? 

Berg:    Well,  its  location.   It  was  known  for  temperatures  in  excess  of  a 
hundred  degrees  Fahrenheit  during  the  summer,  maybe  four  or  five 
months  of  the  year.  And  it  was  a  southern  city  still.   It  was 
relatively  racist.   I  think  blacks  still  had  to  sit  in  the  back  of 
the  bus  at  that  time.   It  was  very  unappetizing.   And  so,  I  told 
Harland  Wood  that  I  would  not  go  to  Cori's  lab;  I  did  not  want  to 
live  in  St.  Louis.   He  was  greatly  disappointed  by  anybody  who 
would  turn  down  an  opportunity  to  work  with  the  great  Carl  Cori. 

Hughes:   You  appreciated  Cori's  stature? 

Berg:    Oh,  I  knew  Cori  by  name,  and  I  had  seen  him  at  meetings.   There 

was  no  question  that  he  and  his  wife,  Gerty  Cori,  both  of  whom  had 
gotten  the  Nobel  Prize,  were  sort  of  the  top  people  in  the  field. 
Arthur  Kornberg  had  gone  to  do  his  apprenticeship  there  and  came 
to  revere  Carl  Cori  as  well.   So  I  lost  a  lot  of  stature  with 
Harland  Wood  by  saying  I  wasn't  going  to  work  with  Carl  Cori.   I 
was  going  to  work  with  Herman  Kalckar  in  Copenhagen  for  a  year 
[1952-1953],  and  then  I  would  spend  a  second  year  [1953-1954]  with 
Arthur  Kornberg  at  the  NIH.   And  that's  what  I  did.   So,  we  went 
off  to  Copenhagen;  I  spent  a  year. 


Postdoctoral  Research  Fellow  with  Arthur  Kornberg, 
Washington  University,  1953-1954 


Berg:     During  the  time  I  was  in  Copenhagen,  Arthur  Kornberg  was  invited 
to  become  the  professor  of  microbiology  at  Washington  University 
in  St.  Louis.   And  so  he  wrote  to  me  and  said  he  hoped  I  still 
planned  to  join  him  when  he  moved  and  took  over  this  new 
department.   And  I  agreed  to  do  it.   So,  I  ended  up  in  St.  Louis 
anyway .   [ 1 aught  e  r ] 

Hughes:   Now,  why  did  you  agree  the  second  time? 

Berg:    Because  the  prospect  of  working  with  Kornberg  seemed  much  more 

appealing  than  working  with  Cori.   Cori  was  a  great  figure;  Arthur 
was  a  rising  young  star.  And  the  kind  of  enzymology  he  was  doing 
was  more  appealing  to  me.   He  was  much  more  aggressive  in  wanting 
me  to  come  than  anything  I  had  ever  heard  from  Cori.   Cori  would 
have  taken  me  largely  because  Harland  Wood  would  have  said,  "I 
want  you  to  take  Paul  Berg  on."  Whereas  Arthur  really  wanted  me 
to  come. 


28 


Arthur  had  gotten  enough  about  me  from  other  people.   I  kept 
in  touch  with  him  when  I  was  in  Copenhagen,  telling  him  about  the 
experiments  we  were  doing.   He  was  very  much  taken  by  the 
discovery  we  made,  and  so  he  wanted  me  to  come.   Also  he  was 
moving  into  a  new  venture.   He  had  always  had  a  sheltered  life 
being  at  the  NIH,  and  here  he  was  now  taking  an  academic  position. 
He  had  the  Job  of  creating  a  whole  new  department.   I'm  sure  he 
must  have  thought  a  little  bit  about  what  kinds  of  people  he 
wanted  to  recruit.   At  least  he  had  a  chance  to  have  a  look-see  at 
me  as  a  postdoc.   And  as  it  turned  out  he  eventually  offered  me  a 
position  in  the  department. 


Postdoctoral  Research  Fellow  with  Herman  Kalckar,  Institute  of 
Cytophysiology ,  Copenhagen,  1952-1953 


Berg:    Copenhagen  was  really  great  because  working  with  Kalckar  was  great 
fun.   I  learned  to  understand  him,  and  if  I  didn't  understand  him, 
I  learned  not  to  be  embarrassed  to  just  keep  plugging  away.   What 
I  found  was  that  the  easiest  way  was  to  take  him  to  a  chalkboard 
or  blackboard,  and  we'd  write  what  we  had  to  communicate.   If  I 
didn't  understand  him,  we  would  try  to  draw  the  experiment.   After 
a  while  I  got  to  be  able  not  only  to  follow  him,  but  almost  to 
anticipate  the  kinds  of  things  he  would  be  thinking  about. 

Hughes:   I  think  of  [Niels]  Bohr- 
Berg:    Bohr  was  almost  as  unintelligible.   In  fact,  I'll  tell  you  a 
little  story.   While  I  was  in  Copenhagen,  Herman  Kalckar  was 
invited  to  speak  at  the  Danish  Royal  Academy  of  Sciences.   He  took 
me  and  one  of  the  other  people  in  the  lab  along  as  his  guests.   It 
was  a  rather  spectacular  Old  World  setting,  with  enormous 
paintings  on  the  wall  of  great  figures  in  Scandinavian  science. 

There  were  two  talks  scheduled,  followed  by  Danish 
sandwiches  and  good  beer.   And  the  first  speaker  was  an  elderly 
man.   In  fact,  most  of  the  members  were  extremely  old.   They  all 
had  earphones,  because  most  couldn't  hear.   I  couldn't  understand 
the  first  speaker;  he  was  speaking  Danish.   It  turned  out  he  was  a 
linguist,  who  gave  a  talk  on  some  of  the  most  erotic  and 
pornographic  passages  in  Danish  poetry.   And  so  there  was  a  lot  of 
snickering,  and  so  on  and  so  forth. 

Hughes:   You  missed  it  all.   [laughter] 

Berg:    And  I  missed  it  all.   I  was  sitting,  looking,  and  staring  around 
at  the  pictures.   Then  Kalckar  got  up,  and  he  started  to  speak. 


29 


He  was  giving  a  scientific  lecture  about  the  research.   And  you 
could  see  within  three  minutes,  five  minutes,  the  earphones  came 
off  all  these  guys,  and  they  sat  back  and  just  either  slept  or 
whatever.   [laughter] 

Bohr,  who  was  there,  fell  asleep.  And  when  he  did  speak,  he 
was,  at  least  to  me,  unintelligible.   I  gathered  that  even  in 
Danish,  Kalckar  was  hard  to  understand.   So  when  he  was  asked  to 
give  lectures  to  the  medical  students,  they  didn't  invite  him  back 
again  after  he  would  give  the  first  couple.   He  had  an  unusual  way 
of  mumbling,  which  is  not  uncommon  amongst  Danes.  And  Bohr 
mumbled  also.   Danes  refer  to  their  language  as  a  throat  disease, 
not  a  language,  plus  this  mumbling.   It  was  just  very  hard.   And 
in  English,  it  wasn't  a  heck  of  a  lot  easier. 

Nevertheless,  we  had  a  great  group  in  the  lab.   It  was  a 
group  from  all  over  the  world—from  India,  from  Canada,  from 
Italy,  and  I  forget  where  else,  plus  Danes,  Swedes.   It  was  a  very 
coherent,  friendly  group.   It  was  an  exciting  period,  because  the 
work  that  was  going  on  in  the  lab  was  booming.   It  was  one  of 
these  things  where  a  new  discovery  had  opened  up  a  whole  new 
world,  and  everything  you  did  was  new  and  successful. 


Discovery  of  Nucleoside  Diphosphokinase 


Berg:    I  got  involved  in  a  project  which  very  quickly  turned  out  to  be 

hugely  successful.   An  Australian  postdoc  Bill  Joklik  and  I  worked 
together  and  published  two  or  three  papers  on  this  one  discovery 
for  one  year's  work. 

Hughes:   Did  you  come  with  an  idea  for  a  research  project? 

Berg:    No.   Kalckar  had  a  physicist  friend  Thomas  Rosenberg  who  dabbled 
in  biology  and  had  some  kind  of  cockeyed  idea  to  explain  a 
particular  reaction  involving  hexokinase.   It's  an  enzyme  that 
converts  glucose  into  a  phosphorylated  form  of  glucose,  and  it's  a 
very  key  step  in  the  metabolism  of  sugars.   It  is  normally  viewed 
as  an  irreversible  reaction.   It's  a  reaction  between  glucose  and 
ATP  [adenosine  triphosphate]  which  forms  a  compound  called 
glucose-6-phosphate  and  ADP  [adenosine  diphosphate] ,  and  was  not 
known  to  be  reversible. 

He  postulated  that  the  reaction  went  in  two  steps;  that  an 
intermediate,  which  was  very,  very  reactive,  immediately  went  on 
to  the  product  that  everybody  knew.   This  intermediate  product,  he 
supposed,  was  a  very  special  form  of  glucose  phosphate,  so-called 


30 


high  energy,  he  called  it.   And  so  we  talked  about  this 
hypothesis;  there  was  no  evidence  for  it.   This  was  a  reaction 
that  Carl  Cori  had  studied  in  great  detail  and  had  found  evidence 
that  there  might  be  a  step  that  was  influenced  by  insulin. 

Rosenberg's  idea  was  that  insulin  involved  the  second  step 
of  this  two-step  reaction.   So,  I  looked  at  it  and  listened.   And 
I  said,  "If  that  is  true,  one  ought  to  be  able  to  do  the  following 
kind  of  experiment  and  show  that  you  could  transfer  P32  from  ATP 
through  this  hypothetical  intermediate  to  another  molecule  like 
ATP,  ITP.   It  was  known  ATP  would  not  transfer  phosphate  to  ITP. 

So,  Kalckar  said,  "That's  an  interesting  idea.   Why  don't 
you  try  and  do  that?"   It  was  known  that  there  was  another  form  of 
ATP,  called  ITP,  which  could  react  with  glucose.   So,  I  came  to 
the  conclusion  that  if  we  took  ATP  with  P32,  and  if  we 
phosphorylated  glucose  to  this  glucose  metaphosphate,  as  we  called 
it,  that  glucose  metaphosphate  could  transfer  the  P32  back  to  ATP, 
but  it  could  also  transfer  it  to  ITP.   So,  in  other  words,  you 
could  transfer  phosphate  from  ATP  to  ITP,  but  it  would  require 
glucose . 

So,  we  set  up  this  experiment.   We  had  to  make  our  own 
radioactive  ATP.   The  idea  was  to  put  in  radioactive  ATP  and  study 
whether  the  P32  transferred  to  ITP  and  required  the  presence  of 
glucose  and  the  enzymes.   Well,  the  experiment  worked  beautifully- 
-P32  went  from  ATP  to  ITP  like  gangbusters,  but  it  didn't  require 
glucose. 

What  we  had  discovered  was  a  new  reaction  in  which  ATP  can 
transfer  phosphate  to  molecules  like  ITP,  and  GTP,  and  CTP,  and 
UTP.   It  doesn't  have  anything  to  do  with  glucose;  it  doesn't  have 
anything  to  do  with  hexokinase.   It  was  a  wholly  new  enzyme.   So, 
just  by  setting  up  an  experiment  to  test  a  particular  hypothesis, 
you  test  one  kind  of  question  and  you  find  something  completely 
different,  which  is  often  true  in  science. 


Making  Radioactive  ATP 


Berg:    There  were  companies  that  were  making  radioactive  ATP,  but  dollars 
were  needed  to  pay,  and  Denmark  didn't  have  dollars.   So,  we  had 
to  make  it  ourselves.   The  traditional  way  of  making  P32-labelled 
ATP  was  to  inject  a  lot  of  P32  phosphate  into  a  rabbit,  exercise 
the  rabbit  so  that  it  breaks  down  all  its  muscle  ATP,  and  then 
allow  the  rabbit  to  recover  and  rebuild  its  ATP.   Then  you  kill 


31 


the  animal  and  you  can  dissect  its  muscles,  and  after  a  whole 
series  of  extractions  and  a  lot  of  complicated  steps  you  get  ATP. 

We  had  an  old  bathtub  up  in  the  attic  of  the  institute.   And 
so  we  decided  we  were  going  to  inject  this  rabbit  with  about,  I 
don't  know,  twenty  millicuries  of  P32,  which  was  a  lot.   We  put 
this  rabbit  into  the  bathtub,  expecting  he  was  going  to  swim,  but 
he  just  sank  to  the  bottom.  We  kept  rescuing  him  and  saying, 
"Swim,  you  bastard."   [laughter]   We  kept  dunking  this  rabbit, 
and  he  just  wouldn't  swim. 

But  eventually  we  got  him  out,  and  he  was  shivering.   And  we 
figured  that  was  enough.   So  we  let  him  shiver  for  a  while,  and 
then  we  wrapped  him  in  a  towel  and  put  him  near  a  radiator  and 
warmed  him  up  for  a  while.   Then  we  sacrificed  him.   Then  we  cut 
him  up  and  isolated  all  his  muscle  and  eventually  isolated  a  lot 
of  ATP.   And  that's  what  we  used.   It  was  such  a  funny  story.   I 
tell  that  to  students  today,  and  they  just  can't  imagine  that 
anybody  ever  made  their  own  ATP. 


Life  in  Denmark 


Berg:    So,  things  went  extremely  well  in  Copenhagen.   We  had  a  great 

time.   We  met  a  lot  of  wonderful  people.   We  traveled  a  lot,  all 
through  Europe.   We  went  skiing  in  Norway,  visited  Sweden, 
traveled  through  Germany  and  Italy  and  the  lakes  region.   It  was 
very  inexpensive.   I  was  living  on  a  fellowship  which  paid  $3,500 
a  year.   We  literally  saved  half  of  it.   We  had  a  villa  in  this 
little  fishing  village  outside  of  Copenhagen,  and  we  saved  enough 
money  to  do  all  this  travel. 

In  fact,  Danish  friends  told  us  we  were  making  more  than  the 
King  of  Denmark,  in  terms  of  our  income  in  dollars.   And  we  ate 
extremely  well,  even  though  Copenhagen  was  just  emerging  from  the 
effects  of  the  Occupation.   The  war  was  over  in  1946;  in  1952, 
there  was  still  rationing  when  we  got  there.   I  remember  going  to 
George  Jensen's  silver  shop  in  the  town,  and  everybody  gathered 
around  the  window  looking,  not  at  the  silver  bowl,  but  at  the 
peaches  it  held. 


Berg:    We  couldn't  buy  certain  kinds  of  food.   Meat  was  rationed.   By  the 
time  we  left,  the  rationing  was  just  coming  off.   I  remember  we 
didn't  have  a  refrigerator  in  this  little  house;  we  had  a  little 
door  to  the  outside  of  the  house  so  milk  could  be  delivered.   The 


32 


milkman  would  open  the  door,  put  the  milk  inside.  It  was  open  to 

the  outside  temperatures;  it  kept  things  cooler.  Eventually  we 

prevailed  on  the  woman  who  owned  the  house  to  get  us  a  small 
refrigerator. 

We  loved  living  there.   We  lived  in  this  village,  which  was 
on  the  edge  of  this  royal  deer  park.   So  when  we  had  to  walk  to 
the  train,  which  took  me  into  work  every  day,  we  walked  along  with 
deer,  and  during  the  mating  season  you  could  hear  this  roaring. 
It  was  incredible. 


James  D.  Watson 


Hughes:   How  common  was  it  then  for  American  graduate  students  to  go 
abroad? 

Berg:     It  was  just  becoming  both  fashionable  and  possible,  largely 

because  of  the  postdoctoral  fellowship  program  that  the  NIH,  the 
American  Cancer  Society,  a  number  of  them  had.   Jim  Watson,  for 
example,  went  to  work  with  Herman  Kalckar. 

Hughes:   Was  that  the  same  year? 

Berg:    Year  before  [1951].   When  Jim  went,  Kalckar  was  not  ever  around. 
Kalckar  had  a  postdoc  whose  name  was  Barbara  Wright,  and  he  and 
Barbara  took  up  with  each  other.   He  left  his  wife,  and  he  and 
Barbara  traveled  all  over  Europe  together.   And  so  Jim  never  saw 
Kalckar.   He  went  to  work  with  Kalckar  to  learn  something  about 
enzymes.   His  advisor  [Salvador]  Luria  said  to  him,  "You  ought  to 
learn  some  chemistry."  And  so  Jim  went  there,  but  Kalckar  wasn't 
there.   So,  he  moved  to  work  with  another  man  named  Ole  Maaloe  who 
was  working  at  the  State  Serum  Institute  on  bacteriophage.   And  to 
this  day,  and  on  many,  many  occasions,  Jim  will  talk  about  this 
lost  year  in  Copenhagen  and  about  Kalckar. 

I  came  the  year  after.   By  that  time  Kalckar  and  his  wife 
had  separated,  and  he  married  Barbara.   And  so  I  got  to  know 
Barbara.   They  had  a  child,  which  I  guess  was  conceived  well 
before  they  were  married.  We  had  a  great  time  together.   So,  for 
me,  that  year  was  very  successful.   It  was  such  a  bad  experience 
for  Watson  that  he  left  Maaloe  after  the  year  and  went  to 
Cambridge. 

Hughes:   Just  think  how  history  would  have  been  different  if  he  had  had  a 
successful  year  with  Kalckar. 


33 

Berg:    He  might  never  have  gone  to  Cambridge.   In  fact,  Watson  tells  the 
story  that  from  Copenhagen  he  went  to  a  meeting  in  Italy  where 
people  were  talking  for  the  first  time  about  DNA  as  the  genetic 
material.   There  was  some  preliminary  structure  work.   And  he  got 
all  turned  on  about  the  need  to  solve  the  structure  of  DNA.   He 
left  Copenhagen  and  got  into  terrible  trouble  with  the  American 
Cancer  Society,  because  they  had  given  him  the  fellowship  to  work 
in  Kalckar's  lab,  and  he  didn't  even  notify  them;  he  just  moved  to 
Cambridge. 

I  remember  Luria  telling  me  that  he  had  to  write  all  these 
letters  to  the  American  Cancer  Society,  trying  to  get  them  to 
accept  that  Watson  was  a  little  bit  bizarre  and  to  forgive  him. 
But  he  was  better  off  in  Cambridge.   And  it  was  in  Cambridge  that 
he  and  Francis  [Crick]  did  that  work  on  DNA  structure. 

Hughes:   So,  he  went  to  Kalckar's  lab  not  with  DNA  on  his  mind  at  all. 

Berg:     No,  not  at  all.   He  wrote  about  that  period  in  his  book  about  the 
discovery  of  the  double  helix.1 


More  on  the  Postdoc  with  Kornberg 
Institutional  Setting 

Berg:    After  Copenhagen,  I  came  back  to  St.  Louis. 
Hughes:   Where  Kornberg  was  already  ensconced.2 

Berg:    He  was  already  there.   He  got  there  in  January  1953;  we  got  there 
in  October.   And  the  lab  was  very  primitive.   It  had  been  an  old 
clinic;  in  fact  it  was  a  clinic  building.   The  first  floor  was 
where  indigent  people  would  be  sitting  or  sprawled  in  chairs  for 
hours,  waiting  to  see  the  doctor.   There  was  this  rickety  elevator 
that  took  you  up  to  the  fourth  floor.   And  there  was  the 
microbiology  department,  which  literally  had  rooms  with  bare  bulbs 
hanging  from  the  roof.   It  was  just  an  amazing  thing. 


1  James  D.  Watson,  The  Double  Helix:  A  Personal  Account  of  the 
Discovery  of  the  Structure  of  DNA,  New  York:  Signet  Books,  1968. 

2  For  Kornberg1 s  views  on  his  time  at  Washington  University,  see  his 
oral  history,  cited  on  page  8. 


Kornberg  took  this  position  because  he  really  wanted  to 
leave  the  N1H.   Cori  had  been  a  very  strong  force  in  persuading 
him  to  come  to  Washington  University.   Besides,  Washington 
University  was  already  well  known;  it  was  a  place  that  had  a 
strong  tradition  in  research.   I  think  Kornberg  must  have  been 
promised  a  whole  lot  of  support.   [phone  interruption] 

Arthur  tells  the  story  that  the  then  dean  of  the  medical 
school,  a  man  named  Oliver  Lowry,  who  was  a  very  well-known 
pharmacologist,  was  head  of  the  pharmacology  department.   He  was  a 
tinkerer.   He  told  Arthur  that  he  would  come  up  and  personally 
paint  the  labs  so  that  they  were  presentable.   He  was  going  to 
help  with  some  of  the  wiring  and  fix  up  the  place.   He  never  did. 
But  he  was  that  kind  of  a  person  anyway.   It  was  so  amusing. 

When  I  got  there,  probably  there  were  two  or  three  labs  that 
were  fixed  up.   One  was  the  one  that  Arthur  and  his  wife  [Sylvy] 
were  working  in,  with  a  couple  of  postdoctoral  fellows.   Another 
one  was  a  very  large  lab  which  could  probably  have  held  six  or 
eight  people,  in  which  there  was  nobody  working  but  had  been  fixed 
up.  Painted,  lit,  it  looked  presentable  as  a  lab.   And  two  other 
labs,  to  which  he  brought  people  who  had  been  postdoctoral  fellows 
at  the  NIH,  and  to  whom  he  gave  appointments  as  professors  in  the 
new  department. 

Hughes:   And  who  were  they? 

Berg:     One  was  named  Osamu  Hayaishi,  who  went  on  to  become  one  of  the 

leading  biochemists  in  Japan  and  today  in  his  eighties  is  probably 
one  of  the  most  influential  people  in  science  in  Japan.   The  other 
was  a  man  named  Irving  Lieberman,  who  eventually  left  never  to  be 
heard  of  again.   I  don't  know  what  ever  happened  to  him.   He  was 
extremely  able  in  the  lab,  but  kind  of  a  weird  guy.   And  the  two 
of  them  never  got  on  very  well,  so  he  left. 

When  I  came,  Kornberg  took  me  into  this  nice  new  lab  and 
said,  "Here's  where  you're  going  to  work."  And  I  looked  around  at 
this  decrepit  place.   He  was  telling  me,  "Don't  worry.   We're 
going  to  get  it  all  fixed  up."  And  we  did  over  the  years.   I  got 
my  Ph.D.  in  "52,  went  abroad  for  the  year,  came  back  from 
Copenhagen  in  '53,  and  immediately  we  talked  about  what  I  might 
work  on.   Kornberg  gave  me  several  suggestions  of  projects. 


35 
Lipmann  and  Lynen's  Hypothesis 

Berg:    While  I  was  in  Copenhagen  there  had  been  a  very  significant  paper 
published  from  Fritz  Lipmann1 s  laboratory- -Lipmann  was  one  of  the 
giants  of  biochemistry  at  the  time—by  a  man  by  the  name  of  Feodor 
Lynen,  who  was  a  professor  in  Munich.   I  can't  remember  whether 
Lipmann  and  Lynen  had  gotten  the  Nobel  Prize  by  then,  or  they  went 
on  to  get  it.1 

Lipmann  and  Lynen  and  two  of  their  students  had  published  a 
paper  which  provided  a  hypothesis  or  model  for  how  a  very  key 
reaction  in  metabolism  worked.   This  was  a  reaction  that  was  very 
central  [acetyl-CoA  formation];  many,  many  people  had  been 
studying  it.   It  was  a  rather  complicated  reaction.   Lipmann  and 
Lienen  proposed  a  novel  mechanism.   A  feature  of  the  mechanism 
that  they  proposed  was  that  the  first  step  was  a  reaction  between 
ATP  and  the  enzyme,  to  knock  off  two  phosphates  from  the  ATP  and 
make  an  AMP  enzyme  complex.   And  then  there  was  a  second  step  in 
which  the  AMP  was  transferred  to  coenzyme  A,  another  one  of  the 
components . 


Berg's  Research  on  Acyl  Adenylates 

Berg:    I  was  intrigued  by  this  notion  of "an  enzyme  intermediate  linked 

with  the  enzyme  AMP.  And  I  was  intrigued  by  it  because  although  I 
was  interested  in  nucleic  acids,  I  didn't  really  know  anything 
about  them.   But  it  seemed  conceivable  that  this  form  of  AMP  was 
an  activated  form  that  might  be  a  precursor  for  building  nucleic 
acids.   So  I  told  Kornberg  that  what  I  wanted  to  do  was  work  on 
this  enzyme-AMP  complex. 

Hughes:   What  was  his  reaction? 
Berg:    He  said  it  was  bunk. 
Hughes:   Why? 

Berg:    Because  the  hypothesis  was  based  on  a  particular  measurement  they 
made,  which  was  to  take  ATP  and  incubate  it  with,  quote,  "an 
enzyme,"  which  was  a  very  murky,  crude  enzyme,  and  show  that  the 
two  terminal  phosphates  would  exchange  with  free  radioactive 
pyrophosphate  in  the  medium.   If  ATP  reacted  with  the  enzyme,  you 


Lipmann  received  the  Nobel  Prize  in  1953. 


36 


would  liberate  inorganic  pyrophosphate  and  produce  a  complex  of 
the  enzyme  with  adenosine  monophosphate  (AMP).   Since  they 
supposed  this  was  a  reversible  reaction,  one  would  anticipate  that 
radioactive  POP  in  the  medium  would  react  with  the  enzyme-AMP 
complex  to  reform  ATP  which  would  be  radioactively  labelled.   And 
that's  what  Lippmann  and  Lynen  found. 

Kornberg  had  already  studied  several  reactions  where  ATP 
exchanged  its  two  terminal  phosphates  with  radioactively  labelled 
pyrophosphate.   But  his  reaction  did  not  occur  by  making  an 
enzyme-AMP  complex,  but  by  another,  more  conventional,  mechanism. 
So  he  said,  "I  don't  believe  any  of  it."   I  said  okay,  that  may 
be,  but  I'm  going  to  take  a  shot  at  it  anyway.   I'm  going  to 
purify  the  enzyme  to  see  if  it  can  produce  the  enzyme  AMP,  and  to 
characterize  it. 

Hughes:  Was  your  enzymology  pretty  good  at  this  point? 

Berg:    Yes,  by  that  time,  I  think  I  knew  how  to  purify  an  enzyme.   I  had 
been  doing  it  in  Copenhagen;  that  was  the  thing  to  do.   We  had 
discovered  a  new  reaction,  isolated  the  enzyme,  purified  it  to 
purity,  and  characterized  that  system.   So,  Kornberg  said,  "Okay, 
if  you  want  to  do  it,  go  ahead." 

Hughes:   Did  he  agree  somewhat  because  you  had  proven  yourself? 

Berg:    Perhaps.   Kornberg  had  a  history  of  directing  the  work  of  people 

in  his  lab.   He  more  or  less  dictated  what  people  did.   So,  it  was 
unusual  that  he  allowed  me  to  do  what  I  wanted  to  do.   It  was  not 
central  to  what  was  going  on  in  the  lab.   But  from  his  estimate  of 
my  ability  or  reputation,  he  let  me  do  it,  even  though  he 
predicted  it  was  going  to  be  a  waste.   And  I  said,  "Well,  we'll 
find  out  if  it's  wrong.   Because  when  I  purify  the  enzyme,  we're 
either  going  to  prove  that  this  is  true  or  it's  not  true."   So,  as 
long  as  I  was  willing  to  approach  it  from  the  point  of  view  of 
purifying  the  enzyme,  that  was  okay. 

The  reaction  I'm  referring  to  was: 

ATP  +  acetate  +  coA  <->  acetyl  coA  +  AMP  +  PPt 

Lipmann  and  Lynen  proposed  that  the  reaction  occurred  in 
three  steps: 


ATP  -  Enzyme  <->  AMP-enzyme  +  PPt 
AMP-enzyme  +  coA  <->  Enzyme-coA  +  AMP 
Enzyme-coA  +  acetate  <->  acetyl  coA  +  AMP 


The  novel  feature  was  the  creation  of  enzyme-AMP  and  enzyme 
coA.   The  evidence  for  their  proposal  was  that  radioactive  PPt  was 


37 


quickly  incorporated  into  the  two  terminal  phosphates  of  ATP  with 
the  enzyme  alone,  none  of  the  other  substrates. 

So,  I  started  off,  and  very  quickly  I  found  that,  as  I 
purified  the  enzyme,  there  was  no  longer  any  exchange  of 
pyrophosphate  with  ATP,  even  though  the  enzyme  was  capable  of 
catalyzing  the  overall  reaction.   I  began  to  ask,  "How  could 
Lipmann  and  Lynen  have  possibly  gotten  those  results?"  These  were 
two  very,  very  distinguished  scientists  who  put  their  name  on  a 
paper  that  was  offering  this  hypothesis. 

And  so  I  began  to  ask,  "Well,  what  if  I  tried  to  carry  out 
this  reaction  not  in  totality,  but  just  parts  of  it?"   In  other 
words,  I'd  leave  out  one  of  the  substrates  so  the  reaction  could 
not  go  all  the  way.   And  what  I  discovered  is  that  when  I  added 
acetate,  one  of  the  other  substrates,  to  the  mixture  of  ATP, 
pyrophosphate,  and  the  enzyme,  it  reconstituted  the  exchange 
reaction.   That  indicated  that  acetate  was  needed  in  addition  to 
the  enzyme  to  promote  the  exchange.   Clearly,  Lipmann  and  Lynen 
were  wrong! 

My  observations  indicated  that  the  enzyme  catalyzed  a 
reaction  between  ATP  and  acetate  to  form  acetyl-AMP. 

ATP  +  acetate  <->  acetyl  AMP  +  PPj 
That  would  make  the  subsequent  reaction: 

acetyl-AMP  +  coA  <->  acetyl-coA  +  AMP 

You  see  that  these  two  steps  produce  the  known  products  but 
not  by  the  three  steps  proposed  by  Lipmann  and  Lynen. 

Hughes:   How  do  you  work  out  a  previously  unknown  molecule? 

Berg:    One  clue  was  the  existence  of  a  somewhat  similar  compound  called 
acetyl  phosphate--m-acetyl  adenylate.   The  most  likely  structure 
was  that  acetate  was  linked  to  the  phosphate  of  AMP.   It  then 
required  that  we  prove  the  existence  of  acetyl-AMP  and  show  that 
it  behaved  in  the  subsequent  reaction  as  indicated. 

So  the  question  is,  how  do  you  prove  the  model?   I  went  to 
the  chemistry  department  and  talked  to  one  of  the  people  there  and 
said,  "If  I  wanted  to  make  something  called  acetyl  AMP,  how  would 
I  do  it?"  People  knew  how  to  make  acetyl  phosphate.   You  just 
took  acetyl  chloride  and  mixed  it  with  silver  phosphate,  and  lo 
and  behold,  you  got  acetyl  phosphate.   So,  I  said,  "Well,  I'm 
going  to  take  silver  adenylate  and  acetyl  chloride,  and  I'm  going 
to  mix  them.  And  I  did.  And  I  got  an  unholy  mixture-mess.   But 
out  of  it  I  purified  something  which  when  I  added  it  to  this 
onzyme,  plus  pyrophosphate,  gave  massive  amounts  of  ATP. 


38 


Then,  working  out  the  rest  of  the  reaction  was  pretty 
simple.   Kornberg  was  always  there  saying,  "Wow,  great,"  and  so 
on.   And  then  there  was  a  national  meeting,  the  Federation  [for 
Experimental  Biology]  meetings,  in  1955,  I  guess  it  was.   I  went 
to  that  meeting,  and  the  word  got  around  of  what  I  had  done.   So, 
I  was  invited  to  give  a  special  presentation,  which  I  did.   And  it 
blew  everybody  away,  including  both  Lipmann  and  Lynen,  who  were 
there. 

Hughes:   What  reaction  did  they  have? 

Berg:    Embarrassment.   Sort  of  a  mixture  of  admiration  and  embarrassment. 
Lipmann  told  me  he  could  never  live  down,  in  his  own  mind,  having 
made  such  a  stupid  mistake.   As  it  turned  out,  they  had  taken  a 
concentrated  extract  of  yeast,  which  was  like  mud,  and  because 
they  had  grown  the  yeast  on  acetic  acid,  it  was  not  surprising 
that  their  extract  contained  some  acetic  acid,  which  was  all  you 
needed  to  trigger  that  reaction. 

Lynen,  in  his  more  sober  moments,  told  me  it  was  the  only 
time  that  anybody  had  shown  him  to  be  wrong.   And  so  for  me  it  was 
kind  of  a  big  high,  because  here  1  was  a  postdoc  and  had  come  on 
to  solve  the  mechanism  of  an  important  biologic  reaction  and 
triumphed  over  Lipmann  and  Lynen.   This  turned  out  to  be  a  very 
general  reaction,  because  all  fatty  acids  are  activated  this  way. 
This  was  the  first  step  in  the  process  by  which  you  build  up  long- 
chain  fatty  acids. 


Rising  Star 

Berg:    So,  there  I  was  in  a  pretty  early  stage  of  my  career  already  with 
two  big  hits.   And  in  publishing  this  work--I  think  there  were 
about  three  papers--Kornberg  did  not  put  his  name  on  them,  which  I 
think  was  a  first  and  last.   I  don't  know  of  any  other  paper  or 
work  that  has  come  out  under  his  tutelage  on  which  he  did  not  put 
his  name.   By  his  not  putting  his  name  on  it,  I  got  the  full 
glory.   Had  his  name  been  on  them,  there's  no  question,  I  would 
not  have  profited  and  benefitted  from  the  discovery  in  the  same 
way  because,  as  is  often  the  case,  the  senior  author  gets  a  lot  of 
the  credit  for  it.  And  the  people  who  did  the  work,  who  actually 
had  the  insight,  or  who  even  came  up  with  the  idea,  get  second 
billing.   I  have  always  been  extraordinarily  grateful  to  him  for 
doing  that. 

I  don't  know  whether  Kornberg  understood  that,  or  whether  he 
just  felt  he  had  not  suggested  that  problem  to  me.   I  had  insisted 


39 


on  giving  it  a  try.   While  he  was  there  for  me  to  talk  to  all  the 
time,  and  was  always  available  for  me  to  bounce  ideas—why  I 
couldn't  get  this  to  work  or  that- -he  in  the  end  felt  it  was  my 
work.   There  was  a  series  of  papers.   I  eventually  described  the 
synthesis  of  this  previously  unknown  compound  and  worked  out  the 
structure. 

So  by  the  time  I  got  to  this  stage,  which  was  1955,  I  was 
pretty  well-known  in  the  field.  And  Kornberg  had  already  asked  me 
to  stay  on  in  the  department.   Even  though  he  didn't  have  a 
faculty  appointment  for  me,  the  school  put  me  up  for  a  Scholar  in 
Cancer  Research  appointment.   It  was  a  special  fellowship  which 
was  to  provide  a  three-year  appointment,  with  the  understanding 
that  the  university  would  give  you  an  academic  appointment  at  the 
first  opening.   But  you  would  be  in  this  interim  position.   It's 
like  the  Markey  Fellowship,  which  serves  to  bridge  the  period 
between  completing  a  postdoc  and  having  the  first  faculty 
appointment . 

Hughes:   [Shows  Berg  his  curriculum  vitae]. 

Berg:     Yes,  "Scholar  in  Cancer  Research,  American  Cancer  Society."   It 
was  for  1955,  and  then  in  '56,  at  the  first  opening,  I  was 
appointed  assistant  professor. 

Hughes:   So  you  worked  independently  at  a  much  earlier  stage  than  is  usual. 

Berg:    Yes,  even  in  graduate  school,  nobody  had  assigned  a  problem  to  me 
for  my  Ph.D.  thesis,  which  is  usually  the  way  things  work.   But  I 
had  suggested  an  idea  of  how  to  solve  a  particular  problem,  so 
that  was  a  kind  of  early  independence.   Even  though  you're  never 
really  totally  independent;  you  get  a  lot  of  feedback  from  people 
all  the  time. 

When  I  went  to  Copenhagen,  I  had  suggested  trying  to  test 
Rosenberg's  hypothesis  in  a  way  that,  as  many  experiments  do, 
leads  you  into  something  that  was  totally  unexpected.   And  then 
coming  to  Kornberg  with  a  preconceived  notion  about  what  I  wanted 
to  work  on,  which  was  different  from  what  was  going  on  in  the  lab 
and  from  what  he  had  suggested  I  might  do.  And  then  this  work  led 
on  to  the  discovery  that  a  similar  kind  of  reaction  happens  with 
amino  acids,  and  that  the  first  reaction  in  the  assembly  of 
proteins  is  to  modify  amino  acids  by  their  attachment  to  AMP  and 
then  be  able  to  attach  them  to  RNA  molecules  and  allow  them  to  be 
assembled  into  proteins.   The  discovery  of  this  form  of 
activation,  as  it  was  called,  turned  out  to  have  great  general 
applicability  and  significance.  And  so,  it  was  seen  as  sort  of  a 
major  discovery. 


Kornberg's  Background 


Hughes:   Not  only  was  Kornberg  an  M.D.  rather  than  a  Ph.D.,  but  he  also  had 
never  had  formal  training  in  biochemistry.   Was  that  a 
consideration  in  terms  of  your  working  with  him? 

Berg:     No,  not  at  all. 

Hughes:   You  looked  at  what  he  had  achieved. 

Berg:    Oh,  yes.   First  of  all,  many  European  scientists,  [Severo]  Ochoa 
and  Cori,  they  were  all  M.D. -trained.   In  Europe,  it  was 
traditional  that  if  you  wanted  to  research  in  the  biological  area, 
you  got  an  M.D.  training.   With  Kornberg,  I  don't  remember  that  I 
even  thought  one  second  about  it,  although  all  the  people  that  I 
had  been  affiliated  with  as  a  graduate  student  were  always 
straight  Ph.D.s.   I  don't  remember  that  it  ever  occurred  to  me  to 
wonder.   In  fact,  almost  all  the  work  Kornberg  did  was  only 
tangentially  related  to  medicine. 

As  a  medical  student,  he  did  research  in  his  own  spare  time, 
and  the  experiments  he  did  were  almost  always  sort  of  biochemical 
in  nature.   He  became  interested  in  nutrition.   He  finished  his 
medical  training,  and  he  went  into  the  coast  guard,  and  he  was 
assigned  to  a  ship.   He  probably  told  you  the  experience  of  trying 
to  take  a  crew  man's  tonsil  out.   They  were  out  at  sea,  and  this 
guy  was  in  terrible  agony  with  this  inflamed  tonsil,  and  Kornberg 
was  the  only  physician  on  board.   So  he  went  in  and  excised  this 
guy's  tonsil,  only  he  was  not  anesthetized.   That  experience 
taught  him  that  medicine  was  not  what  he  wanted  to  do,  and  so  he 
got  transferred  to  the  Commissioned  Corps  of  the  NIH  [National 
Institutes  of  Health],  while  he  was  actually  in  military  service. 
When  he  left  the  Public  Health  Service,  he  was  a  commander,  or 
something,  in  the  navy  rank. 

When  he  went  to  the  NIH,  he  started  working  on  nutrition. 
II 

Berg:    So,  as  Arthur  says,  he  realized  that  there's  all  this  happening 
within  the  animal,  and  you  can't  tell  what's  going  on.   So,  he 
decided  enzymes  were  something  that  he  had  to  learn  about.   He 
went  to  work  with  Cori  and  then  with  Ochoa.   Ochoa  was  a  real 
enzymologist.   And  I  think  Arthur  learned  a  lot  of  enzymology 
during  those  two  stays,  particularly  since  there  were  graduate 
students  and  postdocs  in  the  lab,  and  he  mixed  with  them.   When  he 
went  back  to  the  NIH,  he  was  finished  with  nutrition.  And  I  don't 
know  if  it  was  immediately  after  that  he  became  the  head  of  the 


41 

laboratory  [Chief  of  Enzyme  and  Metabolism  Section,  National 
Institute  of  Arthritis  and  Metabolic  Diseases] .   Soon  after,  there 
were,  in  succession,  spectacular  discoveries  of  new  reactions, 
purified  enzymes,  nailing  down  the  mechanism  of  the  reaction-- just 
one  after  another.   That's  when  [1951]  he  got  this  Paul-Lewis 
Award  in  Enzyme  Chemistry  for  the  enzyme  work. 

I  also  recognized  myself  that  enzymes  were  in  the  end  the 
only  level  at  which  we  could  understand  the  biochemistry,  so  just 
starting  out,  we  realized  we  had  to  get  some  training  and 
experience  with  enzymes.   Kalckar  was  doing  enzymology,  purifying 
enzymes,  measuring  them  spectrophotometrically.   Arthur  was 
purifying  enzymes.   They  were  both  identified  as  rising  stars. 
And  so  an  ambitious  young  guy  is  going  to  definitely  go  to  the 
place  where  the  action  is. 

Hughes:   And  that's  what  the  ambitious  young  guy  did.   [laughter] 


III   RESEARCH  INTERESTS:  NEW  AND  CONTINUING 
[Interview  2:  August  12,  1997]  ## 

Research  on  Fatty  Acid  Activation 

Returning  to  a  Curious  Reaction 


Hughes:   Dr.  Berg,  last  time  we  were  talking  about  St.  Louis,  and  you  have 
more  to  say  about  the  research  that  you  did  there. 

Berg:    The  work  concerned  how  acetic  acid  is  activated  for  its  ultimate 
incorporation  into  long-chain  fatty  acids.   The  mechanism  was 
novel  and  had  not  been  seen  before.   Now,  in  the  course  of 
purifying  the  enzyme  that  carried  out  the  formation  of  acetyl  CoA, 
I  had  come  across  another  reaction,  which  clearly  was  not  that 
system,  but  which  had  the  same  characteristics.   Namely,  it 
catalyzed  the  exchange  of  inorganic  pyrophosphate  with  ATP,  but 
instead  of  acetate  being  the  promoter  of  this  reaction,  it  was  an 
amino  acid,  methionine. 

That  was  curious.   It  wasn't  at  all  clear  why  methionine 
should  be  involved  in  promoting  a  reaction  like  fatty  acid 
activation.   I  put  that  on  the  shelf  and  waited  until  I  completely 
clarified  the  fatty  acid  system.   And  when  that  was  done,  I  went 
back  to  this  reaction  with  the  amino  acid,  and  for  the  life  of  me, 
I  couldn't  figure  it  out.   This  was  a  reaction  with  no  net  charge. 
With  only  ATP,  the  amino  acid  methionine,  and  radioactive 
pyrophosphate,  the  pyrophosphate  would  exchange  with  ATP  very 
rapidly.   Nothing  would  happen  in  the  absence  of  methionine. 

Some  reaction  was  occurring  between  ATP  and  methionine  that 
was  causing  the  release  of  pyrophosphate,  and  that  reaction  was 
reversible,  so  you  could  reincorporate  pyrophosphate  back  into 
ATP.   Well,  I  had  studied  the  fatty  acid  system,  which  had  exactly 
the  same  characteristics,  namely  in  that  case  the  enzyme  uses 


acetate  to  promote  the  exchange  of  ATP  and  pyrophosphate.   Now  the 
acetate  system  has  another  component  to  it,  which  is  coenzyme  A, 
and  coenzyme  A  acts  as  an  acceptor  of  the  activated  acetate.   The 
entire  reaction  involves  first  the  reaction  between  ATP  and 
acetate  to  activate,  and  then  to  transfer  the  activated  acid  to 
acetyl  CoA.   So  I  reckoned  that  the  amino  acid  system  must  be  very 
similar,  that  what  we  were  doing  was  activating  the  amino  acid  to 
some  form,  and  there  had  to  be  an  acceptor.   But  there  was  no 
known  acceptor.   So  the  question  was,  why  don't  we  try  to  find  out 
what  the  acceptor  is?  We  could  easily  devise  an  assay  that  if  you 
had  the  acceptor  the  reaction  would  go  all  the  way,  instead  of 
just  going  back  and  forth. 


James  Ofengand's  Research  on  an  Acceptor 

Berg:    The  first  graduate  student  I  had,  a  fellow  named  James  Ofengand, 
arrived  and  he  was  put  on  that  problem.   I  guess  it  would  have 
been  1955,  because  he  finished  in  four  years  and  left  when  we 
moved  to  Stanford.   So  Jim  started  to  try  to  identify  something 
which  can  act  as  an  acceptor  of  this  activated  amino  acid.   Sure 
enough,  he  fractionated  crude  materials  and  found  something  which 
did  in  fact  act  as  an  acceptor.   And  when  he  ultimately  purified 
it,  it  turned  out  to  be  a  small  RNA  molecule,  which  subsequently 
was  called  transfer  RNA  [tRNA].   And  so  the  reaction  was 
completely  parallel  to  the  fatty  acid  system.   Only  in  this  case 
an  amino  acid  was  being  activated.   It  turned  out  that  the  amino 
acid  was  activated  through  its  carboxyl  group,  just  as  the  fatty 
acid  was.   When  it's  activated  through  its  carboxyl  group,  the 
amino  acid  is  very,  very  efficiently  transferred  to  the  tRNA.   The 
interest  was  that  it  looked  like  it  might  be  involved  as  one  of 
the  intermediates  in  the  assembly  of  proteins. 


Paul  Zamecnik's  Research  on  Amino  Acid  Incorporation  In 
Vitro 


Berg:    There  were  indications  from  other  laboratories  that  amino  acids 

had  to  interact  with  RNA  before  being  incorporated  into  proteins. 
Paul  Zamecnik  at  Harvard  and  Mahlon  Hoagland,  who  was 
collaborating  with  him,  had  been  able  to  show  that  amino  acids 
could  be  incorporated  into  proteins  in  vitro,  and  that  a  molecule 
which  they  called  soluble  RNA  was  required.   It  turned  out  the 
soluble  RNA  was  the  same  as  transfer  RNA  [tRNA].   So,  they  showed 


there  was  an  RNA  requirement  for  incorporating  amino  acids  into 
proteins. 


Purifying  Enzymes  Activating  Some  Amino  Acids 

Berg:    We  found  that  the  amino  acids  were  being  attached  to  this  tRNA, 

and  we  reckoned  that  the  amino  acid-tRNA  was  the  precursor  or  the 
donor  of  the  amino  acid  for  the  assembly  of  protein  chains. 

Hughes:   What  year  was  this? 

Berg:    This  was  1956;  it  could  be  '57,  somewhere  in  that  period.   And  I 
think  our  first  published  works  on  the  activation  of  amino  acids 
is  in  that  period.   Subsequently,  we  went  on  to  show  that  if  you 
looked  back  in  crude  extracts,  you  found  that  there  were  enzymes 
to  activate  different  amino  acids.   By  the  time  we  came  here 
[1959],  we  had  purified  the  enzymes  that  were  necessary  for 
activating  some  six  or  seven  amino  acids,  each  enzyme  being 
specific  for  a  single  amino  acid,  and  each  being  specific  for  a 
separate  tRNA  molecule. 

It  was  our  work  that  really  established  this  notion  of  one 
tRNA  per  amino  acid.   You  could  see  that  there  were  twenty 
different  enzymes;  each  one  of  them  could  recognize  one  of  the 
twenty  amino  acids  to  the  exclusion  of  the  nineteen  others,  and  in 
the  presence  of  ATP  activated  that  amino  acid,  and  then 
transferred  it  to  its  acceptor  tRNA. 

Hughes:  It  sounds  as  though  you  were  not  provoked  by  Watson  and  Crick  and 
the  central  dogma:  DNA,  to  RNA,  to  protein.  You  came  across  this 
by  a  strictly  biochemical  route. 

Berg:    Oh,  yes.   The  central  dogma  at  that  time  was,  DNA  is  responsible 
for  encoding  proteins.   But  the  "DNA  to  RNA"  wasn't  even  known. 
There  was  a  presumption  that  RNA  was  involved,  but  actually  there 
was  a  misconception.   It  was  thought  that  the  RNA  that  was 
involved  was  in  the  ribosomes.   The  ribosomes  are  the  machines  on 
which  the  proteins  are  assembled.   But  in  point  of  fact,  it  isn't 
the  RNA  in  the  ribosomes  which  is  important;  it's  the  so-called 
messenger  RNA.  And  the  messenger  RNA  hypothesis  wasn't  made  until 
1959.   So,  we  were  just  following  our  nose  and  saying,  how  do  you 
assemble  proteins? 

The  Zamecnik  group  was  the  first  one  to  create  an  in  vitro 
system  that  showed  that  if  you  put  in  radioactive  amino  acids, 
they  could  end  up  in  proteins.   If  you  were  a  biochemist  you  would 


say,  "Now,  let's  fractionate  this  system  and  ask  what  are  the 
important  elements  for  that  reaction?" 

We  came  at  it  quite  from  the  back  road.   We  weren't 
interested  in  protein  synthesis.   We  were  fresh  out  of  finding  the 
mechanism  for  activating  fatty  acids  and  stumbled,  if  you  will,  on 
the  fact  that  a  similar  kind  of  system  was  necessary  for 
activating  amino  acids.  And  once  we  recognized  that  enzymes  were 
putting  amino  acids  on  tRNA,  and  knowing  of  the  Zamecnik  work,  we 
were  in  up  to  our  ears  in  protein  synthesis  at  that  point.   So 
then  the  question  to  ask  is,  attaching  an  amino  acid  to  a  tRNA, 
and  each  amino  acid  to  a  different  tRNA,  how  are  all  of  those 
amino  acetyl  tRNAs,  as  they're  called,  used  in  protein  synthesis? 


Mike  Chamberlain  and  Bill  Wood 


Berg:    When  I  came  here  in  1959,  I  had  two  superb  students.   One  was  Mike 
Chamberlain,  who's  a  professor  at  Berkeley,  and  Bill  Wood,  who 
came  from  Harvard;  both  of  them  came  as  Harvard  undergraduates  and 
joined  the  lab.   Bill  Wood  is  a  professor  at  Colorado,  in  Boulder. 

What  Bill  did  was  to  set  up  an  in  vitro  protein  synthesis 
system  in  our  lab.   He  purified  ribosomes,  and  he  prepared  a 
fraction  which  contained  all  of  these  enzymes  and  all  of  these 
tRNAs,  and  demonstrated  that  the  amino  acetyl  tRNAs  were  in  fact 
the  precursor  for  incorporating  amino  acids  into  protein.   Mike 
Chamberlain  was  one  of  the  very  first  people  to  purify  RNA 
polymerase,  because  now  we  knew  that  there  was  a  messenger  RNA. 
The  messenger  RNA  must  have  been  made  off  the  DNA. 

So  what  was  the  enzyme  responsible  for  making  messenger  RNA? 
Mike  Chamberlain,  starting  with  crude  extract  of  E.  coli,  detected 
an  activity  of  converting  ATP,  UTP,  GTP,  and  CTP  into  RNA,  and  he 
began  to  purify  that.   He  was  probably  one  of  the  first  to  have 
actually  published  a  procedure  for  getting  purified  RNA 
polymerase,  and  he  did  a  lot  of  characterization. 

So,  we  had  in  the  lab  then,  after  we  moved  here,  the 
expertise  for  two  parts  of  the  system  for  gene  expression.   One 
was  transcription,  and  the  other  one  was  translation. 
Transcription  generates  the  messenger  RNA;  translation  is  the 
mechanism  for  converting  that  messenger  RNA  sequence  into  a 
protein  sequence. 


46 

Arthur  Kornberg's  Research  on  DNA  Replication 
Research  on  DNA  Synthesis 


Hughes:   If  you  include  Arthur  in  that  picture,  you  have  his  work  on  the 
very  beginning  of  this  process. 

Berg:    On  DNA  replication. 

Hughes:   Arthur  arrived  at  the  DNA  synthesis  problem  circuitously  as  well.1 
His  original  interest  was  not  specifically  in  nucleic  acid 
synthesis,  as  I  understand.   And  you  just  told  me  that  you  arrived 
at  the  problem  of  protein  synthesis  by  a  circular  route. 

Berg:    I  think  ours  was  probably  more  accidental  than  his.   He  has 

written  that,  whether  you  take  it  at  face  value  or  not—it  differs 
among  different  people.  He  says,  "The  fact  that  Watson  and  Crick 
identified  DNA  as  the  genetic  material  had  no  bearing  on  my  choice 
to  work  on  DNA.   I  worked  on  DNA  because  it  was  an  interesting 
biochemical  problem,  and  I  couldn't  have  cared  less  if  it  was  the 
genetic  material."2  I've  wondered  whether  that  was  a  bit 
disingenuous,  but  he  holds  to  that. 

But  it  was  very  clear:  when  he  was  working  on  nucleotide 
synthesis,  he  says  that  he  already  had  in  mind  that  the  long-term 
goal,  if  you  think  the  way  he  does,  was  how  will  these  nucleotides 
assemble  into  nucleic  acids?  So,  in  St.  Louis,  he  was  working  on 
nucleotide  synthesis—how  you  convert  purines  and  pyrimidines  into 
triphosphates .   Already  in  St.  Louis,  and  I  think  it  must  have 
been  something  like  1957,  '58,  he  began  to  ask,  "Now,  if  I  can 
make  radioactively  labelled  nucleoside  triphosphates,  can  I  find  a 
system  that  will  actually  polymerize  them  into  DNA? 

And  so,  the  DNA  synthesis  story  actually  starts  in  St. 
Louis,  and  as  he  has  described  in  his  book,  it's  a  really  an 
extraordinary  story,  because  it  shows  how  a  person  who  is 
determined  will  persist  and  believe  what  most  people  would  not 
have  believed.   In  his  initial  experiments,  the  amount  of 
radioisotope  incorporation  into  DNA  he  got  was  so  small  that  it 
was  barely  above  the  radiation  background.   Many  people  would  have 
ignored  that.   He  maintained  that  the  fifty  counts  above 


p.  8. 


1  See  Arthur  Kornberg's  oral  history,  cited  p.  8. 

2  Dr.  Kornberg  made  a  similar  statement  in  his  oral  history,  cited 


47 


Hughes: 

Berg: 

Hughes: 
Berg: 


Hughes : 
Berg: 


Hughes ; 
Berg: 


background  of  radioactive  material  had  the  characteristics  of  DNA. 
He  could  subject  them  to  various  kinds  of  chemical  tests  and  was 
convinced  that  the  material  he  was  making  in  his  cell-free  system 
had  the  characteristics  of  DNA,  so  he  pursued  it.   He  already  was 
onto  DNA  replication  before  we  left  St.  Louis. 

He  could  have  that  confidence  because  he  was  so  sure  of  his 
technique?   I  mean,  the  fact  that  he  insisted  on  working  with 
clean  enzymes? 


Well,  he  didn't  have  clean  enzymes, 
in  extracts. 

Then  why  was  he  so  confident? 


Those  experiments  were  done 


Well,  you  do  good  controls,  and  you  subject  your  findings  to  the 
most  rigorous  tests.   By  tests,  what  I  mean  is  you  ask:  if  it's 
DNA,  it  should  behave  like  this.   If  it's  not  DNA,  it  can't 
satisfy  all  these  criteria.   So,  you  get  a  little  bit  of  a 
reaction,  and  you  put  this  fairly  intensive  scrutiny  on  it.   And 
if  it  behaves  like  DNA,  you  go  ahead.   And  that's  what  he  did.   In 
fact,  when  he  tried  to  publish  the  work,  it  was  turned  down.   He 
had  one  heck  of  a  time  getting  the  work  published. 

Why? 

I'm  not  sure,  actually.  My  recollection  is  that  it  was  turned 
down  in  large  part  because  calling  it  DNA,  without  showing  it  had 
genetic  properties,  was  sort  of  an  extrapolation.  While  he  had 
something  that  behaved  chemically  like  DNA,  DNA  is  supposed  to  be 
the  genetic  material,  and  he  didn't  have  a  test  for  the  genetic 
activity  of  what  he  had  made.   So,  in  fact  the  paper  kept  being 
turned  down.   Eventually,  it  was  published.   But  the  point  I 
wanted  to  make  was,  he  was  already  on  the  track  to  try  and  learn 
something  about  DNA  synthesis. 

That's  another  reason  why  he  would  pay  attention  to  a  finding  that 
was  barely  above  background. 

Yes,  that's  right.   He  reasoned  that  if  thymidine  was  being 
incorporated  into  DNA,  it  had  to  be  converted  to  something  else 
before  it  could  be  incorporated.   He  suspected  that  the  true 
precursor  must  be  thymidine  triphosphate.   So,  he  set  out  to 
chemically  synthesize  thymidine  triphosphate. 

And  then,  of  course,  the  incorporation  went  much  better. 
The  levels  were  now  much  more  respectable.   He  had  a  good  assay. 
He  could  begin  to  purify  the  enzyme  that  was  incorporating 
thymidine  triphosphate.  As  soon  as  he  began  to  do  a  few 


48 

purifications,  he  showed  it  required  the  other  three  precursors  of 
DNA  (d[deoxy]ATP,  dCTP  and  dGTP).   In  crude  extracts,  they're 
usually  there  in  trace  amounts.   You  can't  show  that  they're 
required,  because  you're  getting  a  very  small  amount  anyway.   But 
once  you  purify  a  little  bit,  you  now  find  that  thymidine 
triphosphate  by  itself  is  not  incorporated;  you  need  all  the 
others.   That  gives  you  more  confidence  that  what  you're  making  is 
in  fact  DNA. 

Hughes:   Yes,  I  see. 

Berg:    So,  it's  kind  of  the  dogged  biochemical  approach,  and  I  think  the 
faith--and  it  is  faith--that  you  can  produce  in  vitro  what  must 
happen  in  the  cell,  and  that  you  have  to  purify  it  to  eventually 
be  able  to  uncover  the  mechanism. 


Nearest  Neighbor  Experiment 


Berg:    Actually,  before  we  left  St.  Louis,  Arthur  had  already  established 
the  parameters  of  that  DNA  system,  to  which  many  people  refer  even 
today.   He  was  able  to  show,  in  what  he  called  the  nearest 
neighbor  experiment,  that  the  frequency  with  which  a  nucleotide 
goes  in  next  to  another  nulceotide  is  exactly  the  same  as  [the 
frequency]  at  which  it  occurs  in  the  DNA  used  as  a  template.   And 
he  proved  some  important  points  about  DNA  replication:  that  it  had 
to  be,  what  we  called,  anti-parallel  synthesis,  which  was  not 
proven  by  the  Watson  and  Crick  structure. 


DNA  as  the  Genetic  Material 


Hughes:   In  the  paper  that  he  had  trouble  publishing,  did  he  show  that  the 
four  bases  were  necessary? 

Berg:    I  think  so. 

Hughes:   And  that  still  was  too  much  of  a  leap  of  faith  to  say,  this  is 
DNA;  this  is  the  genetic  material. 

Berg:    Right.   I  don't  remember  that  the  issue  was  whether  somebody  would 
believe  that  DNA  was  the  genetic  material.   In  fact,  it  was  the 
other  way  around.   It  almost  was  that  the  true  nonbelievers  were 
the  people  who  said,  "Well,  DNA  has  a  property;  it's  the  genetic 
material;  it  confers  genetic  specificity.   How  do  we  know  that 


what  you've  made  is  DNA?   It  was  actually  1967  before  he  actually 
showed  that  he  could  make  a  viral  genome. 

Despite  his  statement  that  he  didn't  care  if  [DNA]  was  the 
genetic  material  or  not,  he  clearly  tried  at  various  stages  to 
determine  whether  the  material  they  were  making  did  have  some 
genetic  property.   They  tried  to  make  transforming  principle,  but 
that  didn't  work.   He  accepted  that  DNA  was  the  genetic  material, 
and  he  was  now  making  it  in  a  test  tube.   The  ultimate  thing  would 
have  been  to  prove  that  what  he  made  in  the  test  tube  had 
biological  activity.  Although,  in  the  beginning  I'm  quite  sure 
that  wasn't  the  motivation. 

So,  Arthur's  getting  into  DNA  replication  was  less 
accidental  than  our  getting  into  protein  synthesis,  which  was 
purely  a  result  of  the  fact  that  we  had  stumbled  onto  a  reaction 
which  resembled  one  which  we  had  been  working  on.   What  got  us 
into  protein  synthesis  was  that  the  reaction  we  found  looked  like 
it  was  producing  the  precursor  to  protein  synthesis, 
[interruption] 


More  on  Amino  Acid  Assembly  and  Messenger  RNA  Research 


Berg:    We  worked  on  that  for  quite  some  time.   Being  the  biochemist  that 
I  was  trained  to  be,  and  under  Kornberg's  tutelage  and 
encouragement,  I  was  pretty  much  a  classical  biochemist  in  his 
mold:  you  find  a  reaction,  you  purify  the  system,  and  you  work  out 
the  mechanism  using  pure  enzymes.   So  what  happened  for  probably 
five  or  six  years  after  we  moved  here  was  to  work  out  this  whole 
system  of  how  amino  acids  become  activated  for  protein  synthesis 
and  the  mechanism  by  which  the  amino  acids  actually  become 
assembled  into  a  polypeptide  chain.   And,  in  addition,  how  you 
make  the  messenger  RNA  that  actually  directs  the  order  of 
assembly.   So  those  were  really  the  major  activities  going  on, 
purifying  all  of  these  so-called  amino  acyl  tRNA  synthetases.   As 
I  say,  there  are  twenty  different  ones. 

You  might  have  thought  the  simplest  scheme  would  be  twenty 
different  tRNA's,  each  one  accepting  only  one  amino  acid.  As  it 
turned  out,  there  are  probably  closer  to  a  hundred  different 
tRNA's,  and  for  any  one  amino  acid  there  are  probably  two,  three, 
or  four  different  tRNA's.  We  came  to  understand  why  that  was 
necessary.   The  genetic  code  has  sixty-four  triplets,  sixty-one  of 
which  actually  specify  amino  acids.   That  means  that  the  code  is 
degenerate,  meaning  there  are  more  codons — triplets  —  for  each 


50 

amino  acid  than  just  one-to-one.   And  if  you  have  different 
triplets  than  you  have  to  have  different  tRNA's. 

What  we  studied  for  some  time  was,  how  does  the  amino  acid 
activating  enzyme  recognize  one  amino  acid  to  the  exclusion  of  the 
nineteen  others?  Because  all  the  amino  acids  have  a  common  core; 
they  have  a  carboxyl  group  and  they  have  an  amino  group,  and  all 
the  amino  acids  look  the  same.   The  only  thing  that's  different  is 
that  they  carry  side  chains  which  are  of  different  kinds.   And 
that  means  that  each  enzyme  has  to  distinguish  each  amino  acid  on 
the  basis  of  its  side  chain. 

Some  of  the  amino  acids  are  very  similar.   It  seemed 
reasonable  to  ask  how  could  one  enzyme  distinguish  between  the 
amino  acids  isoleucine  and  valine,  which  differ  ever  so  slightly. 
You  would  imagine  that  if  an  enzyme  could  recognize  isoleucine,  it 
must  also  be  able  to  recognize  valine.   We  discovered  that  in  fact 
the  enzyme  does  just  that;  it  makes  mistakes.   But  it  has  an 
extraordinary  capacity  to  rectify  its  mistakes,  because  once  it 
transfers  a  wrong  amino  acid  onto  a  tRNA,  it  knows  it  has  made  a 
mistake,  and  it  clips  it  off.   So,  you  never  get  the  wrong  amino 
acid  going  into  a  particular  position  on  a  protein.   We  worked  out 
a  lot  of  the  details  of  that  specificity. 

The  second  part  of  the  specificity:  once  the  enzyme  has  made 
a  particular  amino  acyl  AMP  [adenosine  monophosphate] ,  which  is 
the  intermediate,  how  does  it  recognize  the  correct  set  of  tRNAs 
to  the  exclusion  of  the  hundred  others?  All  the  tRNA's  look  very 
much  alike.   They're  all  small;  they  have  between  seventy  and 
ninety  nucleotides;  they're  all  folded  in  essentially  the  same 
way.   And  yet  the  protein  seems  to  be  able  to  recognize  some  small 
subset,  puts  the  amino  acid  on  those,  and  doesn't  ever  put  it  on 
the  wrong  ones.   So,  we  worked  out  a  lot  of  the  basis  for  that 
specificity,  new  assays  and  so  on  and  so  forth. 

Hughes:   Is  it  a  somewhat  unique  situation  in  biochemistry  that  enzymes  are 
forced  to  be  so  discriminating? 

Berg:    This  is  probably  one  of  the  most  discriminating  of  all.   You  could 
say  polymerase  is  almost  the  other  extreme:  it  uses  any  one  of  the 
four  triphosphates.   And  they  differ.   I  mean,  ATP,  CTP,  and  UTP 
all  look  very  different,  yet  the  enzyme  knows  how  to  use  each  one 
of  them,  but  dictates  which  goes  in  first  or  next;  the  order  is 
determined  by  the  template.   The  enzyme  is  just  there  to  match 
whatever  it  picks  up.   If  it  doesn't  match,  it's  released.   At 
random  it  picks  the  triphosphates;  if  they  form  a  hydrogen  bond, 
it  will  make  the  linkage. 


51 


But  in  the  case  of  amino  acids,  activation  is  very  highly 
specific.   Once  the  amino  acid  is  attached  to  the  tRNA,  its  fate 
is  sealed.   It  will  go  into  a  position  in  the  protein  dictated  by 
the  tRNA,  not  the  amino  acid.   The  amino  acid  could  be  anything. 
Seymour  Benzer  demonstrated  in  a  brilliant  experiment  that  if  you 
put  an  amino  acid  on  a  particular  tRNA,  and  then  chemically  change 
the  amino  acid  (cysteine  into  serine),  it  goes  into  the  protein  in 
position  calling  for  cysteine,  not  serine. 

fl 

Berg:    Thus  producing  the  correct  sequence  in  the  protein  is  determined 

by  the  specificity  of  these  activating  enzymes.   If  the  activating 
enzymes  make  mistakes,  then  the  protein  will  be  incorrect.   That's 
why  we  focused  so  much  on  trying  to  understand  the  nature  of  the 
specificity  and  the  error-correcting  features  that  the  system  has. 
So  between  1959  and  roughly  1965- '66,  we  focused  on  understanding 
both  transcription—the  copying  of  DNA  into  RNA--and  the  assembly 
of  amino  acids  into  proteins,  specifically  on  this  activation 
step. 


Gene  Regulation 


Pasteur  Institute  Contributions 


Berg:    Meanwhile,  of  course,  a  lot  was  happening  working  out  the  holy 
trinity,  which  is  DNA  to  RNA  to  protein.   The  Paris  group  at 
Pasteur  pioneered  in  the  field  of  gene  regulation.   How  do  you 
regulate  whether  a  gene  is  transcribed  or  not? 

It  was  known  for  quite  a  long  time  that  in  normally  growing 
E.  coli,  for  example,  most  of  its  genes  were  not  working.   They're 
only  expressed  when  the  cell  needs  them.   The  induced  enzymes  are 
enzymes  which  are  not  expressed  at  all  unless  the  cell  senses  a 
need  to  make  those  enzymes  in  order  to  metabolize  some  material  in 
the  medium.   Scientists  at  Pasteur  worked  out  a  whole  lot  of  the 
details  of  this  control  mechanism,  involving  so-called  operators 
and  repressers  and  so  on. 

Hughes:   You  had  people  in  your  group  in  St.  Louis  who  had  been  at  the 
Pasteur  Institute. 

Berg:    Melvin  Cohn,  David  Hogness. 
Hughes:   Yes,  and  Dale  Kaiser? 


52 


Berg:    Dale  Kaiser  worked  on  bacteriophage .   He  had  been  to  Pasteur,  and 
he  worked  with  [Andre]  Lwoff  on  a  particular  aspect  of 
bacteriophage  lambda. 


Stanford  Biochemistry  Contributions 


Hughes:   The  idea  of  gene  regulation  was  already  in  the  wind  before  you 
arrived  in  St.  Louis? 

Berg:    Dave  Hogness  and  Mel  Cohn  had  worked  on  that  in  the  late  fifties 
and  had  done  one  of  the  classic  experiments,  which  showed  that 
enzyme  induction  actually  involved  totally  new  protein  synthesis 
rather  than  activation  of  an  inactive  form  of  the  bacterial 
enzyme,  and  induction  is  nothing  more  than  conversion  of  an 
inactive  form  to  an  active  form.   They  showed  that  when  the 
inducer  is  present,  the  cell  begins  to  make  proteins  de  novo,  i.e. 
from  scratch.   And  that  proved  that  induction  was  really  control 
of  protein  assembly. 

Using  genetic  tools  primarily,  [Jacques]  Monod  and 
[Francois]  Jacob  showed  that  there  must  be  a  region  upstream  of 
the  beta-galactosidase  gene  which  regulates  that  gene's  expression 
and  that  there  was  a  represser  that  binds  to  that  regulatory 
region  blocking  gene  expression.   Thus,  when  the  represser  is 
bound  to  that  region  of  the  DNA,  beta-galactosidase  is  not  made. 
In  this  case,  the  inducer  binds  to  the  represser  causing  it  to 
come  off  the  DNA,  allowing  expression  of  the  gene  to  go  on.   The 
details  were  worked  out  I'd  say  in  the  late  fifties  to  early 
sixties  when  Monod  and  Jacob  and  their  colleagues  did  their  famous 
work  identifying  the  operator  (the  control  region)  and  the  operon 
(groups  of  genes  that  are  regulated  together). 

We  were  following  that  work  closely  because  Mel  Cohn  who  had 
worked  with  Monod  was  in  our  department,  and  he  was  educating  us. 
Most  of  us  were  biochemists;  we  were  not  geneticists,  and  we  were 
not  microbiologists,  but  Mel  Cohn  was  both.   And  so  we  were 
learning  about  what  was  going  on  in  Pasteur  through  him.    In 
fact,  Monod  and  Jacob  came  many  times  to  St.  Louis  and  we  got  to 
know  them.   We  became  friends  with  them  as  well  as  discussing 
their  research  with  them. 


53 


Dale  Raiser  and  Bacteriophage  Lambda 


Berg:    My  best  recollection  is  that  sometime  in  1965  Dale  Kaiser  gave  a 
graduate  course  on  bacteriophage  lambda.   He  was  really  already 
one  of  the  experts  in  this  field.  And  he  was  a  wonderful 
lecturer.   The  last  lecture  of  his  course  attempted  to  draw  the 
analogy  between  bacteriophage  lambda  and  a  mammalian  tumor  virus. 

Now  bacteriophage  lambda,  just  to  give  you  a  little 
background,  is  a  virus  which  has  two  possible  outcomes  when  it 
infects  E.  coli.   One  is,  the  virus  multiplies  and  the  cell  dies. 
It  pops  open  and  releases  several  hundred  bacteriophage  particles, 
They  go  on  to  infect  other  cells,  those  cells  lyse,  and  you 
produce  what's  called  the  lytic  phase  of  bacteriophage  lambda 
infections.   That's  the  way  most  bacteriophages  work.   They  kill 
the  cells  in  which  they  multiply. 

But  what  had  been  discovered  in  the  Pasteur  Institute  by 
Andre  Lwoff ,  with  whom  Dale  Kaiser  had  worked,  was  that 
bacteriophage  lambda  frequently  enters  the  chromosome  of  the 
bacterium  and  remains  dormant.   The  bacterium  continues  to 
multiply  perfectly  normally,  but  it  has  acquired  a  new  set  of 
genetic  information,  that  carried  by  the  bacteriophage  genome. 
But  a  complicated  system  keeps  most  of  the  genes  of  the 
bacteriophage  repressed,  and  so  the  bacteriophage  genome  is 
maintained  in  the  bacterial  genome.   But  with  a  very,  very  low 
frequency,  the  virus  genes  are  spontaneously  activated  and  the 
virus  begins  to  multiply  and  kill  the  cells.   The  phenomenon  is 
called  lysogeny. 

Dale  Kaiser  was  one  of  the  experts  of  this  phenomenon,  and 
that's  what  he  had  studied.   Cells  that  are  lysogenic  cannot  be 
infected,  because  they're  producing  a  represser  which  is  keeping 
all  viral  genes  repressed.   So  when  a  new  virus  chromosome  comes 
in,  the  represser  blocks  the  action  of  its  genes,  and  new 
infections  don't  work.   Dale  had  studied  this  phenomenon  called 
immunity.   He  was  a  terrific  colleague.   He  was  bright.   He 
brought  a  whole  new  world  to  most  of  us . 


54 
Research  on  Tumor  Viruses 

Turning  to  Tumor  Viruses  as  an  Experimental  System 


Berg:    Now,  it  was  already  known  that  there  is  a  system  of  mammalian 

viruses  which  infect  cells,  integrate  their  chromosomes  into  the 
chromosomes  of  the  cell,  and  the  cell  doesn't  die.   In  fact,  the 
cell  becomes  transformed  into  a  tumor  cell.   That's  why  these 
viruses  are  called  tumor  viruses.   They  rarely  cause  lysis;  they 
rarely  kill  the  cell. 

SV40  and  polyoma  are  two  viruses  that  are  characteristic  of 
this  group.   They  are  small  DNA  viruses.   When  they  infect  cells, 
depending  upon  the  type  of  cells,  they  either  kill  the  cells  and 
produce  more  virus  particles,  or  they  integrate  into  the 
chromosome  and  remain  dormant.   In  the  latter  case,  they  confer  a 
new  property  on  that  cell,  namely  it  becomes  a  tumor  cell.   When 
Dale  contrasted  these  two  systems,  lysogeny  with  bacteriophage  and 
oncogenesis  or  tumorogenesis  by  tumor  viruses,  he  pointed  out  the 
striking  similarities. 

Hughes:   That  was  a  novel  comparison  at  the  time? 

Berg:    No,  I  think  people  who  had  been  working  with  the  tumor  viruses 

already  were  aware  of  lysogeny  and  were  thinking  that  that  might 
be  a  compatible  system.   But  it  turned  out  not  to  be  the  same 
because  in  the  bacteriophage  a  represser  is  produced  which  blocks 
gene  expression  in  the  viral  genome.   In  the  case  of  the  mammalian 
cells,  there  is  no  represser;  the  virus  DNA  integrates  into  the 
chromosome  and  becomes  stably  associated  with  the  host 
chromosomes.   The  virus  genome  can't  replicate  itself.   But  it 
continues  to  express  some  of  its  genes,  one  or  more  of  which  is 
necessary  to  convert  the  infected  cell  into  a  tumor  cell.   So, 
these  tumor  viruses  carry  a  gene  which  causes  cancer.   As  long  as 
a  cell  retains  that  virus  gene,  it  grows  in  an  aberrant  way.   I 
was  intrigued  by  that.   I  was  getting  a  little  bit  tired  of 
working  with  bacteria. 

I  should  step  back  for  a  moment.   The  studies  with  the 
mammalian  viruses  are  done  in  tissue  culture.   Just  as  in  the 
bacterial  system  you  grow  E.  coli  on  a  petri  plate  and  you  infect 
them  with  a  virus,  you  grow  mammalian  cells  on  a  petri  plate  and 
you  infect  them  with  a  virus,  then  you  can  identify  cells  that 
become  transformed.   I  was  also  well  aware  that  we  had  learned  an 
enormous  amount  about  molecular  biology,  gene  function, 
regulation,  etcetera,  by  studying  the  viruses  that  infect 
bacteria. 


55 


There  were  two  approaches  to  exploring  molecular  biology. 
One  was  to  study  the  bacterial  cell,  and  the  other  one  was  to 
study  what  happens  after  you  infect  the  bacterial  cell  with  a 
virus.   If  you  think  about  it,  when  you  infect  the  cells  a  new  set 
of  genetic  information  is  brought  into  the  cells.   Now  you  have 
almost  a  synchronized  system;  all  the  cells  are  infected,  and  now 
the  virus's  genetic  program  takes  over.   So  if  you  want  to  study 
gene  regulation,  you  could  study  the  regulation  of  the 
bacteriophage's  genes,  rather  than  the  more  complicated  ones  of 
the  cell.   The  viruses  have  a  smaller  genome,  so  there  are  fewer 
genes  to  have  to  worry  about.   And  you  have  this  possibility  that 
you  can  isolate  the  genes  of  the  virus  because  you  can  grow  the 
virus  in  large  quantities.   It's  a  simpler  system,  smaller 
genomes.   So  a  lot  was  learned  using  that  paradigm. 

The  lesson  I  think  that  I  and  a  lot  of  people  drew  was  using 
viruses  as  a  probe  or  as  a  model  often  provided  a  lot  of 
information  about  the  nature  of  genetic  control  and  the  processes 
involved  in  going  from  gene  to  protein.   So,  hearing  that  there 
was  such  a  thing  as  tumor  viruses  and  that  they  induced  cancer  in 
animals  was  intriguing.   But  if  you  look  at  it,  you  would  say  why 
couldn't  you  use  these  viruses  in  the  same  way,  to  study  gene 
regulation  in  mammalian  cells,  rather  than  just  in  bacteria?   I 
used  to  joke  when  I  said  that  what  I  wanted  to  know  was  whether 
the  Jacob-Monod  model  for  regulating  gene  function  was  as  true  in 
mammalian  cells  as  it  was  in  bacteria. 

Hughes:   You  didn't  find  the  complexity  of  the  mammalian  cell  daunting? 

Berg:    Well,  I  didn't,  because  I  was  not  going  to  be  studying  the 

mammalian  cell  genome's  expression  and  regulation.   I  was  going  to 
be  looking  at  the  expression  of  the  viral  genes  after  the  virus 
infects. 

Hughes:   I  see. 


Simian  Virus  40 


Berg:    The  viruses  that  we  chose  to  work  on  had  only  five  genes.   They 
have  a  very  small  DNA  genome;  a  circular  DNA  molecule  of  only 
about  five  thousand  base  pairs. 

Hughes:   Both  SVAO  and  polyoma? 

Berg:    Yes.   SV40  grows  in  human  cells  and  primate  cells;  polyoma  grows 
in  mouse  cells. 


56 


Hughes:   And  their  simplicity  is  at  least  partially  the  reason  they  are 
research  tools? 

Berg:    I  think  the  simplicity  really  only  became  apparent  in  the  mid- 
sixties.   SVAO  was  discovered  in  1961,  inadvertently.   It  turned 
out  that  SVAO  was  a  contaminant  in  the  Jonas  Salk  polio  vaccine. 
The  Salk  vaccine  was  prepared  from  polio  viruses  that  had  been 
grown  on  Rhesus  monkey  kidney  cells.   Virus  was  recovered  in  the 
cell  lysates  without  much  purification  and  then  inactivated  with 
an  agent  that  kills  the  virus  but  doesn't  destroy  its  ability  to 
act  as  a  vaccine.   It  turned  out  that  these  rhesus  monkey  kidney 
cells  were  infected  with  SVAO,  which  nobody  had  ever  known.   SVAO 
stands  for  simian  virus.   It  was  discovered  by  taking  polio  virus 
and  injecting  it  into  hamsters;  much  to  people's  dismay,  the 
hamsters  developed  tumors.   That  was  very  worrisome  because  the 
same  vaccine  was  being  injected  into  kids.   Therein  lies  one  of 
the  things  that  we'll  come  to  in  the  recombinant  DNA  controversy, 
because  we  were  using  SVAO,  and  the  question  was,  does  SVAO 
produce  tumors  in  humans?   It  certainly  produces  tumors  in 
rodents.   The  question  was,  is  working  with  SVAO  dangerous? 


Kornberg's  Dedication  to  E.  coli  as  a  Research  Tool 


Berg:    After  hearing  Dale  Kaiser's  lecture,  I  decided  that  what  I  wanted 
to  do  was  to  stop  working  on  bacterial  systems  and  learn  how  to 
culture  mammalian  cells  and  to  use  SVAO  or  polyoma  as  a  model  for 
studying  gene  expression  and  regulation  of  mammalian  cells. 

Hughes:   Were  your  colleagues  behind  this? 

Berg:    Arthur  was  furious.   I  won't  say  we  ever  came  to  blows,  but  there 
were  times  when  I  was  so  furious  with  him  because  he  was  so 
critical.   He  more  or  less  said,  "You're  wasting  your  talent. 
You're  destroying  your  career.   You  have  so  much  of  a  gift  for 
doing  enzyme  research.   The  only  true  path  to  knowledge  is  E. 
coli,"  and  so  on  and  so  forth.   He  was  very  narrow-minded.   I 
won't  say  he  forbad  me  from  taking  that  step,  but  he  certainly 
predicted  complete  failure.   The  fact  that  we  didn't  fail  and  that 
we  turned  up  a  lot  of  very  important  new  things  about  SVAO,  which 
I'll  come  to,  made  him  even  less  happy.   In  fact,  the  term  he  used 
is  that  I  was  a  Pied  Piper  leading  people  astray,  taking  them  away 
from  important  basic  research  into  this  messy  field  of  working 
with  uncharacterized  systems,  complex  systems  like  mammalian  cells 
and  so  on. 


57 


Arthur  and  I  have  this  debate  to  this  day.   For  example,  if 
you  want  to  study  immunity,  you  can  only  study  immunity  in  a 
system  in  which  immunity  exists.   You  can't  study  immunity  in  E. 
coli.   But  he  won't  concede  that.   He  will  argue  that  principles 
and  models  that  you  learned  from  E.  coli  may  be  the  key  to  unlock 
the  understanding  of  immunity.   For  sure,  but  at  some  point  you 
have  to  work  with  cells  that  display  immunity.   If  you  want  to 
study  oncogenesis  and  tumor  formation,  you  want  to  learn  what  the 
genes  are  that  are  responsible  for  forming  tumors.   You're  not 
going  to  find  out  about  tumor  genes  in  E.  coli.   So,  that's  an 
ongoing  debate.   We  have  it  all  the  time. 

And  we'll  probably  have  it  tonight,  because  we're  having 
dinner  with  Senator  Connie  Mack,  who  is  trying  to  increase  funding 
for  the  NIH.   I'm  sure  that  tonight  at  dinner  Mike  Bishop, 
Kornberg,  and  I  will  get  into  a  debate  about  which  is  the  true 
path  to  knowledge.   [laughter]   Kornberg  will  lament  the  fact  that 
people  like  me  and  Bishop  have  opened  up  vistas  that  have  been 
extremely  informative  and  profitable  in  terms  of  research  and  left 
E.  coli  behind.   He  talks  about  fashions  in  research,  and  the 
fashion  now  is  not  to  work  on  E.  coli.   The  work  which  went  on 
over  the  last  thirty,  forty  years,  will  not  be  used  to  its  fullest 
because  people  are  now  going  off  into  other  research  areas. 

In  fact,  if  I  would  tell  you  some  stories  that  are  really 
amusing:  there  was  a  time  when  I  heard  Arthur  lecture  Gobind 
Khorana  for  leaving  the  work  that  he  was  doing  on  the  chemistry  of 
nucleotides  and  nucleic  acids  to  work  on  membranes,  which  Arthur 
thought  was  a  waste  of  his  talent.   This  was  after  Gobind  had  the 
Nobel  Prize.   I  won't  say  he  berated—that '  s  probably  too  strong 
of  a  word—but  he  certainly  was  furious  with  Francis  Crick  for 
seducing  his  son  Roger  [Kornberg]  to  work  on  chromatin  when  Roger 
went  to  Cambridge  to  the  MRC  [Medical  Research  Council]. 
"Chromatin  was  dirty.   It  was  a  mess.   It  was  too  complex."  Yet 
Roger  made  some  of  the  major  discoveries  that  opened  up  the  whole 
field  of  chromatin  to  a  more  sophisticated  kind  of  study. 

When  I  was  chairman  of  the  department  [1969-1974],  I  had  a 
big  fight  with  him  about  a  professor  in  our  department  who  also 
decided  to  work  on  tumor  viruses  and  went  off  and  spent  a  year  in 
London  at  the  ICRF.   George  Stark  came  back  and  began  to  leave 
what  he  was  doing,  which  was  protein  chemistry,  to  work  on  tumor 
viruses.  Kornberg  read  him  the  riot  act  at  one  of  our  retreats. 
He  just  told  him  he  was  wasting  his  career,  wasting  his  talent.   I 
was  furious,  and  I  told  Arthur  he  had  no  right  to  be  telling 
people  what  to  work  on. 

Anyway,  this  is  a  long  story,  and  an  ongoing  debate  that 
we've  had  over  many  years,  and  continues.   We  differ  over  the 


58 

intensity  of  his  feeling  that  working  on  E.  coli  is  the  only 
worthwhile  system  and  everything  else  is  too  messy.   But  in  fact, 
the  science  of  mammalian  cells  has  really  blossomed. 

Hughes:   I  understand  that  Arthur's  approach  was  a  relatively  common  one, 
probably  up  until  the  late  fifties,  wouldn't  you  think? 

Berg:     Yes. 

Hughes:   The  answers  were  to  be  obtained  by  looking  at  simple  systems,  and 
the  mammalian  cell  was  too  complex  an  entity  and  science  didn't 
yet  have  the  tools  to  approach  it. 


Choosing  a  New  Research  Direction 


Berg:    What  Arthur  didn't  understand,  and  what  I  tried  to  convey  to  him 
on  many  occasions,  was  that  ambitious,  bright,  young  people  want 
to  move  away  from  where  everybody  else  is  working.   They  try  to 
open  new  fields.   If  they  ask  a  question  that  takes  them  in  a 
totally  new  direction,  that's  exciting.   They  want  that  challenge. 

Arthur  was  that  way.   He  was  a  nutritionist.   He  was  working 
on  feeding  mice  or  rats,  and  he  said  he  got  fed  up  with  just 
studying  what  went  in  and  what  came  put  and  not  knowing  what  went 
on  inside.   So,  he  left  nutrition.   Now,  at  that  time  I'm  sure 
somebody  must  have  said,  "You're  going  to  damage  your  career, 
because  you  are  one  of  the  leading  people  in  this  field.   You're 
going  off  to  work  on  purified  enzymes  and  take  a  year  off  to  go 
work  with  Ochoa  and  Cori?"  Well,  when  he  came  back,  he  was  the 
hot,  young  guy  in  a  new  field. 

I  saw  my  position  pretty  much  in  the  same  way.   The 
bacterial  field,  particularly  in  the  area  of  gene  expression,  in 
outlines  were  already  becoming  quite  clear.   The  Jacob-Monod-Lwof f 
story  was  already  the  dominant  paradigm.  And  yet  there  was 
nothing  known  about  gene  expression  in  mammalian  cells,  in 
eukaryotes--nothing!   It  wasn't  a  totally  unnatural  question  to 
ask:   Are  the  mechanisms  that  work  in  bacteria  also  the  ones  that 
will  explain  gene  regulation  in  eukaryotic  cells?  Very  few  people 
were  working  on  that. 

I  was  ambitious,  reasonably  bright,  and  had  already  made  a 
mark  in  traditional  biochemistry.   I  was  really  interested  in 
trying  to  do  something  new,  something  different,  that  other  people 
were  not  doing.  And  I  have  no  doubt,  had  Arthur  been  in  my 
position,  he  would  have  done  exactly  the  same  thing.   I'm 


59 


absolutely  certain.   I  think  that  attitude  has  driven  a  number  of 
other  people  in  our  department  and  made  the  department  so 
successful.   Dale  Kaiser  went  into  working  on  a  totally  new  system 
in  developmental  biology.   Dave  Hogness  who  had  been  working  on 
bacteriophage  X  went  off  and  started  working  on  Drosophila 
genetics.   Each  of  them  have  become  the  leaders  in  the  new  fields 
that  they  literally  created.   Some  people  can  stay  with  what  they 
did  as  graduate  students,  postdocs,  and  follow  along,  and  some  are 
very  successful.   But  for  me,  I  would  get  bored. 


Sabbatical  at  the  Salk  Institute,  1967-1968 


Berg:    When  I  chose  to  work  on  tumor  viruses,  it  was  just  a  budding 

field,  and  one  of  the  leading  people  in  it  was  Renato  Dulbecco. 
Renato  came  up  to  visit  Stanford,  and  I  asked  him  about  the 
chances  of  working  at  the  Salk  Institute.   This  was  because  of 
listening  to  Dale  Kaiser. 

So,  the  question  was,  do  tumor  viruses  express  some  genes 
that  cause  the  cell  to  behave  like  a  tumor  cell?   I  decided  that  I 
was  going  to  take  the  year  off,  learn  how  to  grow  mammalian  cells, 
and  come  back  to  Stanford  and  work  on  tumor  viruses.   That  plan 
did  not  meet  with  Arthur's  approval;  everybody  else  thought  that 
was  great. 

My  assistant  Marianne  Dieckmann  went  with  me  to  La  Jolla.   I 
was  due  to  get  a  postdoctoral  fellow  who  was  coming  from  the 
Pasteur  Institute  to  work  with  me.   When  I  changed  my  plans  to  go 
to  the  Salk  Institute,  he  came  there  with  me.   So,  we  went  down 
there  as  a  small  team--myself ,  Marianne,  and  Francois  Cuzin,  who 
had  come  from  Francois  Jacob's  lab  at  the  Pasteur  Institute.   We 
started  working  with  tumor  viruses,  adapting  the  strategies  from 
molecular  biology.  All  the  concepts  that  we  had  of  regulation,  of 
the  way  bacteriophage  multiply,  how  they  lysogenize,  we  brought 
with  us  to  the  animal  virus  field.  When  we  came  back  to  Stanford, 
I  was  committed;  that  was  the  new  direction  we  were  going  to  go 
in.   And  the  work  that  was  ongoing  with  the  amino  acid  activating 
enzymes  sort  of  tailed  off. 


Establishing  Research  on  SV40  at  Stanford 


Berg:    I  was  away  during  '67- '68,  and  when  I  came  back  I  built  a  new 
laboratory,  which  had  all  the  safety  features — filtered  air, 


60 


negative  pressure,  laminar  flow  hoods,  and  everything  of  that 
sort.   The  point  is  I  was  committed;  I  was  going  to  work  in  that 
field.   We  did  some  important  work  of  opening  up  a  whole  field: 
finding  mutations,  characterizing  the  genome  of  SV40--it's  a  small 
circular  DNA  molecule,  5243  gene  pairs—and  trying  to  determine 
which  regions  of  the  DNA  specified  which  gene.   We  worked  out  a 
whole  lot  of  new  technology  for  doing  that.   It  was  built  on 
methods  that  we  brought  with  us  from  the  earlier  work  with 
bacteriophages,  which  is  usually  the  way  science  progresses. 


Building  on  the  Bacteriophage  Lambda-£.  coli  Research 


Hughes:   Your  concept  was  that  what  happens  in  E.  coli  infection  is  a  basis 
for  thinking  about  what  happens  in  mammalian  cells? 

Berg:    Absolutely.   For  example,  the  virus  chromosome  is  a  small  DNA 
molecule;  it  has  to  be  transcribed;  the  cell's  transcription 
machinery  resembles  that  of  bacteria  in  having  an  RNA  polymerase 
which  it  uses  the  triphosphates ,  and  so  on.   So  what  you  know 
about  transcription  in  E.  coli,  you're  going  to  immediately  apply 
to  transcription  in  eukaryotes.   It  was  possible  to  map  the  RNAs 
and  to  characterize  them,  that  is,  when  they're  made  after  the 
infection.   As  it  turns  out,  when  the  virus  enters  the  cell,  part 
of  its  genome  is  transcribed  into  what  we  call  early  messenger 
RNAs.   Those  early  messengers  make  early  proteins. 

Those  early  proteins  then  start  the  system  of  replication, 
and  you  then  begin  to  express  a  new  set  of  genes,  so-called  late 
genes.   The  late  genes  encode  the  shell  of  the  virus,  very  similar 
to  that  of  the  bacteriophage.   Bacteriophage  lambda  T-4  do  almost 
the  same  thing:  they  enter  E.  coli  and  express  part  of  their 
genome,  early  genes;  they  make  early  proteins;  early  proteins 
start  replication.   The  replication  initiates  late  transcription 
which  results  in  the  proteins  that  form  the  phage.   Very  similar, 
right?  But  now  you've  opened  up  a  whole  new  world  of  how  a 
mammalian  cell  does  that.  And  you  can  begin  to  study  the 
mechanism  by  which  mammalian  cells  carry  out  the  basic  steps  of 
gene  expression. 


Drawbacks  of  Using  Commercial  Reagents 


Hughes:   What  difference  does  it  make  that  students  today  come  to  mammalian 
cell  research  without  the  experience  that  you  and  others  of  your 


61 


generation  had?  Do  they  lose  something  because  of  not  having 
experience  with  phage  and  E.  colil 

ii 

Berg:    I  think  the  big  difference  today  is  students  are  unaware  of  the 
amount  of  work  it  takes  to  prepare  all  of  their  own  reagents. 
When  Kornberg  was  studying  DNA  replication,  he  had  to  synthesize 
all  the  radioactive  deoxytriphosphates.   To  the  people  in  his  lab, 
it  was  like  being  sentenced  to  Siberia  when  it  was  their  turn  to 
take  several  months  out  and  prepare  the  labeled  triphosphates. 
Today,  students  just  write  an  order  and  buy  it.   They  buy  all 
their  enzymes;  they  don't  purify  any  enzymes—ever .   And  when  they 
buy  them,  they  have  no  respect  for  their  use.   They  never  worry 
about  how  much  activity  there  is;  they  just  dump  in  some  amount. 
The  manufacturer  says  use  five  microliters;  they  don't  know  what 
five  microliters  contain.   They  don't  know  if  the  enzymes  have 
impurities  and  so  on  and  so  forth. 

Those  of  us  who  grew  up  in  a  more  traditional  mode  worry 
about  all  these  things.   If  you  get  a  result  that's  funny,  maybe 
it's  funny  because  your  enzyme  preparation  is  contaminated,  not 
because  there's  some  funny  phenomenon.   But  people  who  have  moved 
into  the  animal  field,  who  work  with  Drosophila,  are  very 
sophisticated. 


Biochemical  and  Genetic  Approaches 


Berg:     Paramount  today  is  the  genetic  approach.   In  working  on  the  Beadle 
biography,1  I've  come  to  recognize  the  power  of  the  approach  which 
he  exploited,  which  in  fact  even  preceded  him.   If  you  have  a 
complex  biological  phenomenon  or  system,  you  can  approach  it  in 
two  ways.   Kornberg 's  approach  is  to  break  open  the  cell  and  try 
to  identify  some  reaction  you're  interested  in,  and  purify  the 
components  responsible  for  the  reaction.   The  other  approach  is  to 
make  mutants—ones  which  block  the  phenomenon,  obliterate  the 
phenomenon,  change  the  phenomenon,  or  prevent  the  reactions  that 
you're  interested  in  studying  from  happening. 

The  various  steps  that  are  involved  in  this  process  are 
dissected  by  making  mutations  in  each  and  every  one  of  the  steps. 
Consider  a  complex  pathway  in  which  a  very  simple  precursor  is 


1  At  the  time  of  the  interviews,  Dr.  Berg  and  Dr.  Maxine  Singer  were 
collaborating  on  a  biography  of  George  Beadle. 


62 


converted  via  ten  steps  to  an  end  product,  and  each  step  is 
catalyzed  by  an  enzyme.   You  could  try  to  purify  all  of  those  ten 
enzymes  and  work  out  the  detailed  chemistry  of  each  step.   Or  you 
could  start  by  making  mutations  which  identify  the  steps,  because 
when  you  make  a  mutation  that  blocks  a  step  in  the  process  the 
substrate  of  that  step  accumulates.   Because  they  accumulate,  the 
intermediates  in  the  reaction  can  be  identified. 

Having  mutants  provides  a  powerful  tool  to  look  for  the 
protein  or  the  genes  which  encode  the  protein.   If  you  have  cells 
that  can't  carry  out  one  of  the  steps  in  the  pathway,  you  can  now 
use  such  cells  to  isolate  the  gene,  because  introducing  that  gene 
into  these  cells  overcomes  the  block.   These  are  the  two  different 
paradigms  for  trying  to  analyze  complex  biological  systems. 

Mutations  are  the  predominant  approach  when  we  have  no  idea 
what  reactions  are  involved  in  the  complex  system  under  study.  Fly 
behavior  is  a  good  example,  for  example,  phototropism.   Normal 
flies  move  to  a  light  source,  and  it  is  possible  to  obtain  mutants 
that  fail  to  do  that.   You  get  mutants  that  can't  see,  mutants 
that  can't  fly,  and  mutants  that  can't  translate  the  light  signal. 

This  [genetic]  paradigm  is  what  people  are  doing  today. 
It's  no  less  honorable  than  purifying  enzymes.   Some  enzymologists 
put  their  faith  in  enzymology,  but  I  suppose  I'm  more  liberal.   I 
admire,  accept  and  encourage  people  who  want  to  try  new  ways  to  do 
things  and  don't  insisting  that  purifying  enzymes  is  the  only  way. 


Fluid  Disciplinary  Boundaries  and  Multidisciplinarity 


Hughes:   Is  it  difficult  to  label  what  people  in  the  biochemistry 

department  actually  are?  Are  they  biochemists,  geneticists,  or 
cell  biologists? 

Berg:    I  think  that's  happening  in  all  areas.   In  the  Pharmacology 

Department,  you  couldn't  distinguish  two- thirds  of  the  people  over 
there  from  the  people  in  my  department.   Similarly,  in 
Microbiology,  people  are  doing  the  same  kinds  of  things  that  are 
being  done  in  Biochemistry.   The  artificial  definitions  don't 
prevail  anymore.   Many  scientific  problems  have  now  gotten  to  be 
very  complex  and  really  need  multiple  and  different  kinds  of 
approaches  for  their  solutions. 

One  of  my  jobs  here  at  the  Beckman  Center  is  to  create 
mechanisms  that  bring  people  together  from  different  departments, 
ones  who  bring  different  perspectives,  different  skills,  different 


63 


technologies  to  a  problem.   The  ability  to  solve  complicated 
problems  rarely  exists  in  any  one  department  any  longer.   The 
problems  are  too  complicated.   So,  what  we're  trying  to  do  is  to 
create  interdepartmental  programs  around  themes,  around  areas  of 
interest. 

Kornberg  tends  to  resist  these  new  approaches.   Arthur 
remains  a  strong  advocate  for  the  enzymological  approach,  using 
pure  enzymes.   He  can  admire  new  approaches,  but  he'll  still  say, 
"You  don't  really  understand  it  until  you  get  the  enzymes  out  and 
purify  them."  That's  good.   It  serves  a  very  important  function. 
You  can  make  the  argument  that  the  purpose  of  the  mutational 
approach  is  to  identify  the  genes  responsible  for  the  individual 
steps.   Ultimately,  however,  to  understand  that  pathway,  you  must 
purify,  identify,  and  characterize  the  proteins  that  perform  those 
steps.   Nobody  denies  that. 

One  time  when  we  were  recruiting  a  new  faculty  member, 
Arthur  and  I  interviewed  a  young  woman  who  had  already  done  some 
really  fantastic  work  at  the  MRC  [Medical  Research  Council]  in 
Cambridge,  using  the  nematode  as  a  model  for  studying  development. 
We  brought  her  over.   She  gave  a  brilliant  seminar.   Kornberg 
asked  her  why  she  wanted  to  be  in  a  biochemistry  department  if  she 
was  a  developmental  biologist.   She  explained  that  the  approach 
she  was  using,  which  was  to  obtain  mutants,  was  only  to  get  into 
the  system  and  that  ultimately  she  wanted  to  be  able  to  understand 
these  mutations  at  the  biochemical  level.   Then  Arthur  asked  her, 
"Why  should  the  biochemistry  department  be  interested  in  you,  a 
developmental  biologist?"  And  she  said,  "I  didn't  ask  to  come; 
you  invited  me  to  come  here."   [laughter]   She  was  pretty  good. 
She's  at  UCSF  now  and  one  of  their  real  stars. 

Hughes:   Who  is  that? 

Berg:    Her  name  is  Cynthia  Kenyon.   She's  now  one  of  the  leaders  in  the 
nematode  field  and  using  nematodes  to  study  aging.   She's  been 
making  mutants  whose  life  spans  are  much  longer  than  normal. 
Interestingly  enough,  Arthur's  son  Tom,  at  UCSF,  is  using 
Drosophilia  mutants  to  study  their  embryonic  development.   So 
there  you  are. 


Research  Leading  to  Recombinant  DNA  Work 
Hughes:   Let's  discuss  your  recombinant  DNA  work. 


Berg:    A  reasonable  starting  point  is  to  ask  how  did  we  ever  get  to 
thinking  about  recombinant  DNA  while  working  on  SVAO? 

In  moving  from  one  field  to  another,  you  bring  to  it  some 
background,  some  insight  and  some  prejudices,  if  you  will.   As  I 
was  preparing  to  go  off  on  sabbatical,  I  was  getting  more  and  more 
involved  with  using  genetic  tools  to  study  how  amino  acyl-tRNAs 
work  in  protein  synthesis. 


Collaborating  with  Charles  Yanofsky  on  Suppressive  Mutations 


Berg:    One  of  my  colleagues  in  the  biology  department,  Charles  Yanofsky, 
and  I  became  very  close  friends,  and  we  began  to  collaborate  on 
certain  experiments.   He  had  demonstrated  that  certain  kinds  of 
mutations  could  reverse  the  effects  of  other  mutations;  some 
rautational  suppressors  appeared  to  work  through  mutations  in 
tRNAs.   That  is,  if  a  tRNA  is  supposed  to  read  a  codon  A-B-C,  you 
can  get  a  mutation  in  the  gene  that  specifies  this  tRNA  so  it  now 
reads  A-B-D,  and  not  A-B-C.   As  a  consequence,  a  particular  amino 
acid  will  be  inserted  into  the  protein  chain  in  the  wrong  place. 
Such  mistakes  can  reverse  the  effects  of  a  mutation  in  the  protein 
coding  gene;  hence  the  term  suppressor. 


Using  Phage  as  Transducing  Agents 


Berg:    We  were  studying  mutations  which  affect  tRNAs  that  change  the 

reading  or  the  translation  of  the  genetic  code.   In  order  to  do 
that,  I  was  beginning  to  learn  to  do  a  lot  of  genetic  crosses  with 
bacteria  and  using  bacteriophage  for  what  we  call  transducing 
agents.   Lambda  is  one  of  a  whole  series  of  phages  which,  after 
integrating  into  the  chromosome  and  then  coming  out,  incorporate 
pieces  of  chromosome  into  their  own  chromosomes. 

Hughes:   By  mistake? 

Berg:    More  or  less  by  mistake.   So,  they  integrate  at  a  particular 
place,  and  with  some  low  frequency,  they  come  out,  but 
incorrectly,  picking  up  a  piece  of  bacterial  DNA  while  losing  a 
bit  of  their  own.   When  such  virus  particles  infect  another  cell, 
what  do  they  do?  They  bring  with  them  the  information  that  they 
stole  from  the  first  cell.   So,  you  can  transfer  genes  from  one 
cell  to  another  by  bacteriophages .   And  that  proved  to  be  a  very 


65 

powerful  technique  in  altering  the  genetics  of  cells  by  bringing 
in  genes  from  different  sources. 

Hughes:   Could  you  specify  the  gene  to  be  carried? 

Berg:     Yes,  because  some  bacteriophages ,  like  lambda  and  another  one 

called  phiSO,  integrate  in  only  one  place  in  the  bacterial  genome. 
So  when  they  come  out,  they  can  only  bring  out  the  pieces  that  are 
alongside.   There  are  other  phages,  however,  that  cause  the 
bacterial  genome  to  be  fragmented  into  small  pieces,  and  when  the 
phage  is  assembled,  it  picks  up  cellular  DNA  pieces  at  random. 
The  pieces  it  picks  up  are  roughly  the  size  of  its  normal  genome, 
and  the  particle  looks  like  a  bacteriophage,  but  it  has  a  piece  of 
bacterial  DNA  instead  of  phage  DNA.   The  pieces  are  picked  up  at 
random.   Consequently,  some  phages  are  carrying  one  gene,  another 
phage  is  carrying  a  different  gene,  another  phage  is  carrying  a 
different  gene,  and  so  on. 

This  notion  that  viruses  could  carry  genes  from  one  cell  to 
another  really  was  a  reality,  and  I  was  using  it  all  the  time. 
It's  called  transduction.   So  phage  could  transduce  cells  with  a 
variety  of  genes  from  any  cells  in  which  the  phage  was  grown. 


Developing  a  Transducing  System  for  Mammalian  Cells 


Berg:     Well,  while  1  was  working  on  the  tumor  viruses,  the  question  I 
asked  myself  was,  are  mammalian  viruses  capable  of  picking  up 
mammalian  viruses  and  bringing  them  into  new  mammalian  cells?   In 
other  words,  could  you,  in  fact,  develop  a  transduction  system 
that  works  for  mammalian  cells,  just  as  the  bacteriophage  work 
with  bacterial  cells?   The  reason  for  believing  that  was  possible 
is  that  when  I  was  at  the  Salk  Institute,  phage  particles  that 
carried  cellular  DNA  were  discovered.   The  question  was,  could 
those  viruses  that  are  carrying  cellular  DNAs  be  used  to  transform 
other  cells?  After  making  some  calculations,  it  became  quite 
clear  that  SV40  and  polyoma  couldn't  do  it,  because  their  genomes 
were  very  small.   Their  genomes  comprise  only  five  thousand  base 
pairs,  and  you  can't  pack  more  into  the  virus  particle.   So,  the 
most  you  could  possibly  pick  up  from  a  mammalian  cell  would  be 
five  thousand  base  pairs. 

When  bacteriophage  picks  up  cellular  genes,  it  loses  a 
little  bit  of  its  own.   But  lambda  is  fifty  thousand  base  pairs; 
SVAO  is  only  five  thousand.   So,  the  question  is,  how  much  could 
you  lose,  and  how  much  could  you  actually  pack  in?  The  answer  was 
that  the  amount  you  could  pack  in  to  the  virus  particle  is  hardly 


66 


big  enough  for  a  single  gene.   And  even  if  it  was  big  enough  for  a 
single  gene,  the  likelihood  that  any  particle  contains  the  gene 
you're  interested  in  or  knew  how  to  look  for  was  problematic,  like 
looking  for  the  needle  in  a  very  large  haystack. 

I  knew  that  we  had  to  find  a  way  to  move  genes  from  one  cell 
to  another.   We  thought  that  if  we  couldn't  find  virus  particles 
that  had  picked  up  a  specific  cellular  DNA,  then  why  not  make 
them?  We  wondered  if  we  could  we  take  a  set  of  pure  genes  and 
insert  them  into  the  SV40  genome  in  vitro.   If  we  could,  then  we 
might  use  the  virus  DNA's  ability  to  enter  a  mammalian  cell, 
integrate  into  the  chromosome,  and  carry  with  it  whatever  had  been 
attached  to  it. 

Hughes:   The  reason  that  you  were  interested  in  the  DNA  of  SVAO  was  simply 
as  a  transporting  mechanism? 

Berg:     Absolutely.   That  was  the  notion  we  had.   If  we  could  create  such 
a  system,  it  would  really  greatly  enlarge  the  capability  of 
studying  mammalian  cells  and  doing  molecular  and  cell  biology.   A 
question  was,  what  could  we  attach  to  SV40? 

At  that  time  in  Dale  Kaiser's  lab,  Ken-ichi  Matsubara  was 
working  with  a  small  piece  of  bacterial  DNA  that  could  replicate 
in  E.  coli.   It  was  part  lambda  bacteriophage  DNA  associated  with 
three  genes  from  E.  coli.   This  plasmid  could  be  purified  and 
obtained  in  pure  form.   The  plasmid,  called  lambda  dv  gal,  was 
about  five  thousand  base  pairs  in  length  and  contained  three 
bacterial  genes  and  the  little  piece  of  lambda  DNA  which  allowed 
it  to  replicate  in  £.  coli.   We  had  pure  SV40  DNA,  and  we  knew 
that  SVAO  DNA  could  be  used  to  infect  mammalian  cells,  whereupon 
new  viruses  are  produced,  or  the  DNA  integrates  into  the  cells' 
chromosomes. 

We  had  pure  SV40  DNA  which  contains  about  five  thousand  base 
pairs  in  the  form  of  a  ring.   We  thought  that  if  we  could  make  a 
molecule  which  contains  SV40  DNA  as  well  as  the  bacterial  genes, 
we  could  ask  if,  after  introduction  into  a  mammalian  cell,  the 
bacterial  genes  are  expressed.   That  was  the  question  we  set  out 
to  answer. 


Lambda  Bacteriophage  with  Complementary  Tails 


Berg:    First  we  had  to  figure  out  how  do  you  join  two  DNA  molecules.   A 
colleague  at  Stanford,  Dale  Kaiser,  had  been  studying  a  lambda 
bacteriophage ' s  cohesive  or  sticky  ends.   The  phage  DNA  has 


67 


single-strand  protrusions  from  each  end  which  allow  molecules  to 
join  end-to-end.   Under  certain  conditions,  lambda  DNA  could  join 
end  to  end  to  make  long  chains  or  under  other  conditions  they 
circularize.   So  the  existence  and  behavior  of  cohesive  ends  in  a 
lambda  phage  DNA  pointed  the  way  to  do  the  joining.   If  you  make 
complementary  tails  on  DNAs,  those  DNA  will  join  to  one  another 
through  the  formation  of  hydrogen  bands. 

Hughes:   You  mean  literally  make  the  tails,  synthesize  them? 

Berg:    Phage  lambda  DNA  has  them  naturally.   What  Dale  Kaiser  and  Al 

Hershey  and  a  few  other  people  all  discovered  simultaneously  was 
that  these  ends  are  like  Velcro.   They're  cohesive,  because 
they're  complementary  to  each  other.   And  so  under  the  appropriate 
conditions,  a  linear  molecule  of  lambda  will  form  circles,  or 
[join]  end  to  end  to  produce  long  chains. 


Synthesizing  "Sticky  Ends" 

Berg:    So  we  set  about  to  learn  how  to  synthesize  synthetic  sticky  ends, 
because  the  two  DNA  molecules  we  had  did  not  have  cohesive  ends. 
Once  you  were  aware  of  the  fact  that  cohesive  ends  exist  naturally 
and  allow  DNA  molecules  to  join  one  to  another,  you  would  say  the 
trick  is  to  make  synthetic  ends.   Because  strands  of  A's  and  one 
with  T's  pair,  it  seemed  logical  to  put  tails  of  A  on  one  [DNA 
molecule]  and  tails  of  T  on  the  other,  and  after  mixing  them,  they 
very  likely  would  join.   So,  the  task  became  one  of  creating 
sticky  ends  with  an  enzyme. 


DNA  Ligase 


Berg:     Now  most  of  the  enzymes  for  doing  this  whole  operation  were  in 

Kornberg's  refrigerator,  and  we  had  access  to  all  of  them,  which 
was  one  of  the  great  things  about  the  department. 

Hughes:   There  were  several  people  involved  with  isolating  ligase,  not  all 
of  them  at  Stanford. 

Berg:  Three  labs  get  credit  for  discovering  the  enzyme  DNA  ligase 
virtually  simultaneously.  Bob  Lehman,  Kornberg,  and  Martin 
Gellert. 

Hughes:   If  you've  got  base  pairing,  where  does  the  ligase  come  in? 


68 


Berg:    Normally,  SV40  DNA  is  a  closed  circle,  that  is,  there  are  no  nicks 
or  gaps;  each  chain  is  absolutely  continuous.   If  you  circularize 
a  linear  molecule  as  I  described  it,  a  DNA  ligase  is  needed  to 
join  all  the  ends.   In  the  case  of  lambda  DNA,  when  you 
circularize  it,  there's  perfect  fit  right  up  to  where  you  make  the 
ligase  join  the  ends  creating  the  closed  circles.   With  uneven 
tails  of  A's  and  T's,  circularization  occurs,  but  these  are  gaps. 
These  have  to  be  filled  in  before  ligase  can  join  the  ends. 


Recombinant  DNA 


Making  Recombinant s  of  SV40  and  Lambda  dv  gal 


Berg:     Making  synthetic  tails  was  the  first  step  [sketching]  [See  diagram 
A,  page  68a].   Let's  see,  these  are  A's,  and  these  lengths  are 
variable  because  we  had  no  way  of  controlling  their  lengths.   With 
dATP  and  an  enzyme  (deoxynucleotidyl  transferase)  A's  are 
polymerized  onto  the  ends.   We  try  to  put  on  somewhere  in  the 
range  of  a  hundred  to  a  hundred  and  fifty  A's  per  end.   But  the 
two  tails  are  certainly  not  the  same  length.   Now  T's  are  added 
with  dTTP  and  the  same  enzyme.   When  the  two  DNAs  are  joined, 
there  are  four  gaps  because  the  dA  and  dT  tails  are  not  all  the 
same  length. 

Hughes:   I  see  that. 

Berg:     To  fill  in  the  gaps  and  seal  the  ends  we  used  DNA  polymerase  I, 
the  Kornberg  enzyme,  which  fills  in  the  gaps,  and  DNA  ligase  to 
seal  the  ends.   All  we  had  to  do  was  create  the  cohesive  ends, 
anneal  them,  add  DNA  polymerase  and  ligase,  and  covalently  closed 
circles  would  be  formed,  one  half  of  which  would  be  SV40,  and  the 
other  half  lambda  dv  gal. 

Hughes:   That's  very  clear. 
Berg:    So,  it's  very  simple. 


Peter  Lobban's  Research  on  Recombinant  DNA 


Berg:    Actually,  Peter  Lobban,  who  was  a  graduate  student  in  Dale 

Kaiser's  lab,  had  come  up  with  the  same  idea  on  his  own.   One 


68a 


AAAAA 


AAAAA  +  TTTTT- 


-TTTT 


Paul  Berg:  "Arrows  denote  gaps  to  be  filled  in  before  DNA  ligase  can  close 
the  circle." 


69 


could  have  used  G's  and  C's  for  cohesive  ends,  but  he  also  used 
A's  and  T's  on  each  end.   What's  necessary  is  that  they  be 
complementary,  that  is,  form  base  pairs  and  join  the  two  DNAs  to 
one  another.   So  this  was  what  we  developed  in  1970-71,  "69. 


The  Jackson,  Symons ,  Berg  Paper,  1972 


Berg:    A  postdoctoral  fellow  in  my  lab,  who  had  done  his  graduate  Ph.D. 
work  here  in  biology  with  Yanofsky,  came  over  and  joined  my  lab 
and  actually  did  this  experiment.   And  it  took  not  more  than  about 
seven  or  eight  months. 

Hughes:   Who  was  that? 

Berg:  David  Jackson.  And  there  was  a  sabbatical  visitor  from  Australia 
named  Bob  Symons.  So,  the  paper  is  actually  Jackson,  Symons,  and 
Berg.1 

Hughes:  If  you  were  to  single  out  one  paper,  would  that  be  the  paper  upon 
which  the  Nobel  Prize  was  based? 

Berg:     Yes,  I  think  so.   If  you  read  the  paper,  you  see  that  in  the 

discussion  it  lays  out  that  the  ability  to  join  two  DNA  molecules 
together  allows  one  to  begin  to  make  all  kinds  of  recombinants. 
We  used  lambda  dv  gal,  but  any  piece  of  DNA  would  work.   We 
anticipated  that  some  technology  would  evolve  in  the  future  to 
isolate  individual  genes,  and  therefore  individual  genes  could  be 
plugged  in. 

Lambda  dv  gal  plasmid  itself  has  the  capacity  to  replicate 
in  E.  coli.   So  if  this  molecule  is  introduced  into  E.  coli,  it 
could  be  replicated  and  maintained  as  a  plasmid.   If  put  into 
mammalian  cells,  we  presumed  that  it  would  integrate.   As  it 
turned  out--Stan  Cohen  was  right—we  opened  up  the  lambda  dv  gal 
at  a  position  that  destroyed  its  ability  to  replicate.   And 
therefore  this  molecule  would  not  have  replicated  in  E.  coli. 

Hughes:   How  did  you  interpret  that  at  the  time? 


1  D.  A.  Jackson,  R.  H.  Symons,  and  P.  Berg,  "Biochemical  methods  for 
inserting  new  genetic  information  into  DNA  of  Simian  Virus  40:  Circular 
SV40  DNA  molecules  containing  lambda  phage  genes  and  the  galactose  operon 
of  Escherichia  coli,"  Proceedings  of  the  National  Academy  of  Sciences 
[PNAS]  1972,  69:2904. 


70 
Berg:     We  didn't  because  we  didn't  try  to  do  it. 

Complexity  of  the  Berg  Recombinant  DNA  Method 

Berg:    Now,  this  technique  of  making  recombinant  DNAs  was  viewed  as  being 
complicated  and  probably  beyond  the  ability  of  most  labs.   It  was 
assumed  that  it  could  be  done  only  in  my  lab,  probably  because  we 
had  all  the  enzymes  locally  and  the  expertise  in  how  to  use  them. 
And  that  was  certainly  correct. 

Once  we  decided  that  we  were  not  going  to  try  to  propagate 
these  recombinants,  our  critics  breathed  a  sigh  of  relief  and 
said,  "Okay,  we've  been  spared.   If  Berg  is  not  going  to  put  these 
recombinants  into  bacteria,  there's  no  longer  a  problem." 

Hughes:   Other  people  wouldn't  be  able  to  do  it. 

Berg:     Couldn't  do  this,  right.   David  Hogness,  one  of  my  colleagues,  was 
the  only  person  who  actually  used  this  (method]  to  construct 
recombinants  with  Drosophila  DNA. 

The  Cohen-Boyer  Recombinant  DNA  Cloning  Method 

Berg:     But  the  whole  picture  changed  when  it  was  discovered  that  when 
certain  restriction  enzymes  cleave  DNA  they  create  natural 
cohesive  ends. 

Hughes:   So  now  practically  anybody  could  do  it. 

Berg:    Now  anybody  could  do  it.   You  could  buy  an  enzyme,  take  two  DNAs, 
cut  them,  mix  them,  tie  them  together  and,  presto,  a  recombinant 
DNA. 

Hughes:   I  gathered  from  your  MIT  interview  that  Asilomar  I  was  originally 
conceived  as  a  two-step  conference.1  But  the  idea  of  having  the 
second  conference  was  temporarily  dropped.   It  was  only  when  the 


1  Interview  with  Paul  Berg,  by  Rae  Goodell,  May  17,  1975,  Recombinant 
DNA  Controversy  Oral  History  Collection,  Institute  Archives,  MIT, 
Cambridge,  p. 28-29.   Berg  was  interviewed  again  for  MIT,  by  Charles  Weiner, 
on  April  17,  1978. 


71 

Cohen/Boyer  experiments  were  disclosed  that  the  need  to  consider 
their  safety  implications  arose. 

Berg:    The  implications  of  the  Cohen/Boyer  cloning  and  the  ease  with 

which  recombinants  could  be  made  was  taken  up  by  a  small  group  of 
seven  people  that  met  at  MIT  in  April  of  1974.   That  meeting 
resulted  in  the  so-called  Berg  letter,1  or  the  moratorium  letter. 


Recombinant  DMA  Controversy 

Concern  about  Berg's  Proposed  Experiment  with  SV40 


Berg:    We  didn't  ever  try  to  put  the  lambda  dv  gal-SV40  hybrid  into  E. 
coli  because  when  it  was  discovered  that  we  were  making  this 
molecule,  there  was  concern  about  putting  SVAO  into  a  bacterium 
that  inhabits  people's  intestines.   The  concern  was  that  SVAO 
carries  tumor  genes,  and  cancer  might  be  spread  through  this  kind 
of  infection. 

So,  reports  of  our  work  in  1971- '72  created  a  big  furor. 
Nick  Wade  wrote  a  book  called  the  Ultimate  Experiment.2  The 
ultimate  experiment  was  to  put  the  SV40-containing  hybrid  plasmid 
into  E.  coli.   We  decided  not  to  do  that  because  we  couldn't  be 
sure  that  this  would  not  produce  a  bacterium  which  could  get  out 
of  the  lab  and  infect  people  and  possibly  populate  their 
intestinal  tract  with  bacteria  that  carried  cancer  genes.   So,  we 
never  did  the  experiment.   We  never  tested  whether  it  could  grow 
in  bacteria. 


Hughes:   One  of  the  debates  was,  could  bacterial  genes  not  only  replicate 
but  also  express  protein  in  mammalian  cells? 


1  "Potential  biohazards  of  recombinant  DNA  molecules,"  Science  1974, 
185:303  (July  26,  1974).   The  signers  of  the  letter  were:  Paul  Berg,  David 
Baltimore,  Herbert  Boyer,  Stanley  Cohen,  Ronald  Davis,  David  Hogness, 
Daniel  Nathans,  Richard  Roblin,  James  Watson,  Sherman  Weissman,  and  Norton 
Zinder. 

2  Nicholas  Wade,  The  Ultimate  Experiment:  Man-made  Evolution,  New 
York:  Walker  and  Co.,  1977. 


72 

Berg:    That's  right.   That  was  why  we  did  the  experiment.   We  planned  to 
put  this  plasmid  into  mammalian  cells.   That  was  not  a  problem;  we 
could  have  done  that,  but  that  was  put  off  because  Dave  Jackson 
left  the  lab  for  a  job  at  the  University  of  Michigan. 
Furthermore,  concerns  arose  and  expanded  about  the  safety  of 
working  with  these  DNA  molecules  themselves.   Being  infectious, 
could  such  molecules  escape  into  the  air  and  get  into  bacteria? 
These  really  were  very  absurd  kinds  of  concerns. 


Berg's  Involvement 


Berg:    But  the  fact  is  that  I  got  distracted  and  got  involved  in  this 

whole  debate  about  whether  these  molecules  were  safe  or  not.   As  a 
consequence,  a  lot  of  people  who  were  leaving  bacterial  work  to 
work  on  tumor  viruses  raised  the  question  of  whether  SV40, 
polyoma,  and  adenovirus  were  a  hazard  for  man. 

In  response  to  these  concerns,  I  got  involved  in  organizing 
a  conference--Asilomar  I  [  1973] '--that  most  people  don't  know 
about.   It  was  held  at  least  two  years  before  the  Asilomar 
Conference  on  Recombinant  DNA  [February  1975]  to  consider  possible 
hazards  of  working  with  tumor  viruses. 


Biosafety  at  Stanford  and  the  Salk  Institute 


Berg:    Remember,  I  mentioned  that  when  I  came  back  from  Salk,  I  built  a 
new  laboratory,  equipped  with  the  latest  biosafety  equipment, 
because  there  was  concern  on  the  part  of  people  in  my  department 
who  were  working  outside  my  group  about  whether  they  were  being 
unduly  exposed  to  potentially  tumor-causing  viruses. 

Hughes:   Did  those  safety  measures  pertain  at  Salk? 

Berg:    Salk  was  very  loose.   They  had  just  these  little  Plexiglass  hoods, 
and  you  did  all  your  work  in  there.   Everybody  was  working  on 
SV40,  and  they  didn't  worry  about  it  at  all.   Generally, 


1  The  publication  arising  from  this  conference  is:  Biohazards  in 
Biological  Research,  Proceedings  of  a  Conference  held  at  the  Asilomar 
Conference  Center,  Pacific  Grove  California,  January  22-24,  1973,  A. 
Hellman,  M.N.  Oxman,  and  R. Pollack,  eds.,  Cold  Spring  Harbor  Laboratory, 
1973. 


73 


scientists  didn't  worry  about  whether  working  with  SV40  was 
dangerous  to  them.   But  then  you  bring  it  into  an  open  lab,  with  a 
lot  of  people  who  are  not  involved  in  the  work.   I  don't  remember 
exactly  how  the  word  got  around  and  why  the  technicians  and 
dishwashers  at  Stanford  would  have  been  worried  about  somebody 
working  with  SV40.   Perhaps  it  was  at  a  departmental  meeting.   I 
had  just  become  chairman  [1969-1974]  of  the  department,  and  so  I 
was  very  sensitive  to  this  kind  of  unrest  and  uncertainty.  To 
mitigate  these  concerns,  we  built  a  lab  with  filtered  air, 
negative  pressure,  laminar  flow  hoods,  in  an  adjacent  building. 
Everybody  was  reasonably  confident  it  was  safe. 


Asilomar  I  Conference,  1973 


Berg:    Out  of  this  conference,  for  which  there  was  a  big  report  and 

book,1  there  was  a  recommendation  to  draw  blood  every  six  months 
from  everybody  working  in  a  lab  and  to  determine  if  they  had 
antibodies  as  a  measure  of  their  exposure  to  the  virus  they  were 
working  with.   If  you  had  antibodies,  it  meant  you  were  exposed  to 
the  virus  and  had  been  infected.   As  it  turned  out,  in  my  lab 
almost  everyone  became  seroconverted. 

Hughes:   That  procedure  arose  from  the  first  Asilomar  conference? 

Berg:    Yes.   [Indicates  photomicrograph  on  book  cover]   That's  SV40  on 
the  cover,  from  my  slides.   [The  arrangement  of  the  virus 
particles]  happened  to  form  a  question  mark. 

Hughes:   Yes,  isn't  that  striking. 

Berg:    That's  the  proceedings  of  the  conference,  the  papers  that  were 
presented  and  the  final  recommendations.   I  made  the  closing 
remarks.   Essentially  what  it  says  is,  we  don't  know  whether  these 
viruses  are  oncogenic  for  man,  and  asks  what  steps  should  be  taken 
to  protect  ourselves.   We  had  in  mind  the  problem,  How  do  we 
proceed  in  the  face  of  uncertain  safety?  Do  we  ignore  it,  or  do 
we  try  to  do  something? 

Not  too  many  people  know  about  the  Asilomar  I  conference 
proceedings.   For  example,  here  are  designs  of  the  kinds  of  hoods 


1  Biohazards  in  Biological  Research,  Proceedings  of  a  Conference  held 
at  the  Asilomar  Conference  Center,  Pacific  Grove,  California,  January  22- 
24,  1973,  A.  Hellraan,  M  .N.  Oxman,  &  R.  Pollock,  eds.,  New  York:  Cold 
Spring  Harbor  Laboratory,  1973. 


74 

that  laboratories  should  construct.  We  brought  in  people  who  were 
in  charge  of  biosafety  at  the  NIH,  and  they  described  how  rooms 
should  be  arranged  and  so  on  and  so  forth—facilities  and 
equipment  available  for  virus  containment. 

Hughes:   Was  there  overlap  of  the  people  who  attended  Asilomar  I  and  II? 

Berg:    Oh,  yes.   There  were  many  of  the  same  people  who  are  in  here 

[referring  to  Asilomar  I  book].   Here's  a  chapter  on  laboratory 
hazards  from  aerosols.   Those  people  were  at  Asilomar  II,  or  they 
were  on  the  RAG  [Recombinant  DNA  Advisory  Committee],  or  the 
guidelines  called  on  them  to  provide  information  about  safety. 
Now  that  I  look  at  this  list  of  attendees,  a  good  many  of  the 
people  were  also  at  the  Asilomar  II  conference. 

One  of  the  steps  we  proposed  was  to  initiate  a  prospective 
study,  to  periodically  collect  blood  samples  from  people  who  were 
working  in  the  field  and  store  them.  We  still  have  them,  frozen 
away.   The  purpose  was  to  keep  track  of  anybody  who  developed 
cancers  and  to  check  with  their  prior  exposure.   Working  with 
laminar  flow  hoods,  in  negative  pressure  labs  were  also  amongst 
the  recommendations  that  came  out  of  this  conference. 

These  procedures  set  the  stage  for  entering  into  an  era 
where  we  were  going  to  work  with  infectious  organisms  whose 
complete  properties  couldn't  be  predicted.   Clearly,  as  we  began 
to  manipulate  viruses  and  make  mutants,  there  was  a  possibility 
that  we  might  make  something  that  would  be  more  dangerous  to  the 
workers  as  well  as  to  the  people  around  us. 


The  Moratorium  Letter  and  the  Meeting  at  MIT 


Berg:     Some  of  the  people  who  were  at  the  MIT  meeting  had  also  been 
participants  in  the  Asilomar  I  meeting. 

Hughes:   Do  you  remember  who  they  were? 

Berg:    Richard  Roblin  was  one;  Jim  Watson,  Dave  Baltimore,  Norton  Zinder, 
were  also  at  Asilomar  I. 

Berg:    The  reason  Roblin  was  invited  to  the  meeting  was  because  he  had 

written  an  article  in  Science  about  gene  therapy  which  focused  on 
manipulating  mammalian  cells  by  virus  infections.  So,  he  was 
invited  to  come  to  that  meeting. 

Hughes:   Why  was  he  provoked  to  write  that  article? 


75 


Berg:    Well,  the  whole  notion  of  genetic  engineering  of  animals  was  not 
new.   Josh  Lederberg  had  speculated  on  it  earlier.   There  were  a 
number  of  science  fiction  scenarios;  in  fact,  oftentimes  gene 
therapy  was  cited  as  justification  for  doing  genetics. 

Hughes:   Arthur  Kornberg  testified  in  the  Senate  in  1968  on  genetic 
engineering. ' 


Andy  Lewis  and  Natural  Adenovirus-SVAO  Recombinants 


Berg:    A  man  named  Andy  Lewis,  working  with  adenovirus  at  the  NIH,  had 

raised  concerns  about  adenovirus-SV40  recombinants .   Adenoviruses 
are  big  viruses  of  about  thirty  thousand  base  pairs.   What  Lewis 
found  is  that  cells  that  were  coinfected  with  SV40  and  adenovirus 
produced  recombinants  which  had  replaced  part  of  the  adenovirus 
with  SVAO  sequences. 

These  were  called  adeno-SV40  hybrids,  and  they  propagated  as 
viruses.   But  they  were  defective;  generally,  when  adenovirus 
genes  are  replaced  by  SV40  genes,  the  recombinant  virus  can't 
multiply.   But  Lewis  discovered  a  class  of  adeno-SVAO  hybrids 
which  were  nondefective  and  could  replicate.   He  was  very 
concerned,  because  we're  all  infected  with  adenovirus  and  if  SV40 
got  out,  it  might  form  adeno-SVAO  hybrids  which  could  propagate 
and  spread  SV40  genes.   Consequently,  a  very  big  part  of  the 
Asilomar  I  meeting  addressed  the  concern  about  what  to  do  about 
these  adeno-SVAO  hybrids. 

Hughes:   What  would  be  the  purpose  in  an  evolutionary  sense  for  SV40  and 
adenovirus  to  be  able  to  join? 

Berg:     There's  no  disadvantage  or  advantage.   It  happens  accidently. 
Hughes:   That  doesn't  happen  very  often,  does  it? 

Berg:    No.   But  here  they'd  cloned  these  out;  they  had  pure  populations 
of  these  adeno-SVAO  hybrids.   People  found  them  very  useful  and 
wanted  to  use  them  as  tools. 

Hughes:   So  you  were  creating  an  artificial  circumstance. 

Berg:    That's  right.   Andy  Lewis  to  this  day  is  concerned  about  such 
events.   There  was  just  a  conference  at  the  NIH,  "Is  SV40 


See  the  oral  history  with  Arthur  Kornberg,  cited  p.  8. 


76 


Tumorigenic  for  Man?"  Because  a  number  of  people  have  come  down 
with  tumors  with  SVAO  genes  in  them.   They're  saying  it  just  took 
a  long  time  for  this  to  happen.   One  of  the  sessions  at  this 
recent  meeting  was  called  "Hazards  Associated  with  Modern  Research 
Methodologies . " 


James  Watson's  Stances 


Berg:    Coming  out  of  the  Asilomar  I  conference,  Jim  Watson  decided  to 

forbid  the  use  of  feline  leukemia  viruses  or  cats  at  Cold  Spring 
Harbor  because  he  was  so  worried  about  whether  these  viruses  could 
cause  cancer  in  man. 

Hughes:   Yet  one  of  the  amazing  things  that  happened  at  Asilomar  II  was 
that  Watson  changed  his  tune  about  the  dangers  of  research  with 
tumor  viruses. 

Berg:    That's  right.   If  you're  cynical,  you  might  speculate  on  why.   Jim 
was  at  the  MIT  meeting  that  came  out  with  the  Berg  letter.   He  was 
absolutely  supportive,  insisting  that  we  had  a  responsibility  to 
warn  the  general  public  and  scientists  about  the  potential  dangers 
of  cloning. 

One  of  the  consequences  of  the  Asilomar  II  conference  was  to 
mandate  that  research  could  proceed  only  under  conditions  which 
guaranteed  the  organisms  you  were  working  with  would  not  escape 
from  the  lab.   I  had  already  invested  in  building  a  P3  [physical 
containment  3]  laboratory  that  minimized  that  risk,  and  it  was 
expensive.   Jim  was  now  pushing  working  with  tumor  viruses,  SVAO, 
at  Cold  Spring  Harbor,  and  he  realized  that  if  such  safe 
facilities  were  required  he  would  have  to  invest  and  build  much 
secure  facilities  there.   Jim  could  certainly  calculate  what  the 
cost  would  be  to  build  the  kind  of  facilities  being  proposed  at 
Asilomar,  and  he  knew  that  not  doing  so  would  impede  their  ability 
to  compete.   I  suspect  he  also  came  to  the  conclusion  that  we  had 
been  rash  at  the  first  meeting  in  presuming  risks  that  he  now 
thought  were  unreasonable. 

Hughes:   At  one  point  you  accosted  him  over  his  change  in  stance.   Can  you 
remember  what  he  replied? 

Berg:    Jim  never  would  have  admitted  that  there  was  an  economic  component 
to  his  argument,  or  his  ability  to  compete.   I  think  what  he  said, 
and  he  may  have  felt  that  honestly,  is  we  made  am  mistake  in  the 
Berg  letter  by  concluding  that  there  was  great  concern. 


77 


Hypothetical  Risks 


Berg:    I  think  if  you  reflect,  as  we  all  did  later,  on  the  basis  for  this 
concern,  it  was  all  hypothetical.   There  was  not  strong  reason  to 
believe  that  what  we  were  doing  would  be  dangerous.   It  sounded 
dangerous.   We  wondered  what  might  be  the  consequences  if  you  put 
genes  that  confer  resistance  to  antibiotics  into  bacteria  that 
infect  man;  you'd  prevent  the  use  of  antibiotics  that  cure 
whatever  that  bacteria  caused.   That  sounds  pretty  worrisome,  and 
we  suggested  that  such  experiments  should  not  be  done.   Second, 
putting  genes  that  specify  toxins  into  bacteria  that  could  inhabit 
man  should  also  not  be  done.   But  then  when  you  come  down  to  the 
rest  of  it,  it  was  pure  hypothesis.   We  could  imagine  you  might 
inadvertantly  pick  up  oncogenes  from  mammalian  DNA,  incorporate 
them  into  plasmids,  and  put  them  into  bacteria.  Well,  so  what? 
As  it  later  turned  out,  even  if  you  do,  it  doesn't  make  any 
difference.   It  turns  out  it's  safer  to  work  with  oncogenes  that 
way  then  it  is  to  try  to  work  with  the  viruses  which  carry  them. 

Hoof  and  mouth  disease  is  one  of  the  serious  virus 
infections,  so  all  work  on  hoof  and  mouth  disease  is  done  on  Plum 
Island,  an  island  off  the  coast  of  Long  Island.   But  you  can 
fragment  hoof  and  mouth  disease  [virus]  and  clone  its  genes,  and 
it's  quite  safe  in  that  form.   Hepatitis  can  only  be  grown  as  DNA 
segments,  as  plasmids.   We  began  to  realize  that  in  some  ways  you 
could  say  it  was  safer  to  be  using  recombinant  DNA. 


Nonmicrobiologists  and  Research  on  Infectious  Organisms 


Hughes:   Asilomar  II  was  February,  1975.   You  began  working  with  SV40  in 

1971? 

Berg:     Nineteen  sixty-nine. 

Hughes:   Do  you  remember  when  Lewis  began  working  with  adeno-SV40  hybrids? 

Berg:     Yes,  it  was  around  '71. 

Hughes:   So,  by  Asilomar  II,  there  was  five  or  six  years  of  research 
experience  with-- 

Berg:    Not  with  recombinant  DNA,  since  the  Cohen/Boyer  experiments  were 
done  during  '73,  '74. 


78 


Hughes:   I'm  calling  what  you  had  done  recombinant  DNA,  and  in  a  sense, 

Lewis's  research,  even  though  I  know  that  the  adeno-SVAO  hybrids 
were  not  a  deliberate  creation.   What  I'm  trying  to  assess  is  how 
much  experience  you  collectively  had  had  with  recombinant 
organisms  by  Asilomar  II.  And  it  seems  to  be  roughly  six  years  of 
research. 

Berg:    Right.   But  only  a  very  small  number  of  people  had  had  that 
experience.   Remember,  the  [Cohen-Boyer]  recombinant  DNA 
breakthrough  made  it  possible  for  anybody  to  do  anything .   This 
was  recognized,  and  was  one  of  the  driving  forces  for  the 
moratorium  letter.   There  was  a  lot  of  people  who  had  no 
experience  working  with  potentially  pathogenic  organisms  and  they 
would  be  moving  into  the  field.   In  fact,  it  was  common  for 
biochemists  and  molecular  biologists  who  had  been  working  with  E. 
coli  to  grow  up  E.  coli  in  five-gallon  jugs  and  dispose  of  some 
down  the  drain.   E.  coli  was  viewed  as  innocuous,  and  some  people 
might  have  volunteered  to  drink  it.   But  microbiologists  and 
bacteriologists  knew  that  E.  coli  was  not  innocuous. 

Everybody  was  pipetting  viruses  by  mouth;  that's  the  way  we 
worked.   The  reason  people  who  worked  with  viruses  were 
seroconverted  was  because  when  you  suck  up  a  column  of  a  solution 
with  a  virus  in  it,  you  have  a  vapor,  an  aerosol  at  the  top,  and 
that  gets  taken  up  [in  the  digestive  track]. 

We  realized  that  in  fact  the  technology  now  had  changed  the 
way  people  were  going  to  do  things,  and  most  of  those  who  were 
going  to  be  working  in  the  field  were  totally  inexperienced  and 
unaware  of  the  most  trivial  safety  measures.   And  so  part  of  the 
rationale  for  the  letter  was  to  bring  this  to  people's  attention. 

Hughes:   I  know  from  talking  to  microbiologists  that  they  had  scathing 
remarks  to  make  about  the  laboratory  safety  techniques  of  non- 
microbiologists . ' 

Berg:    Microbiologists  used  a  loop.   I  don't  think  a  biochemist  knew  what 
a  loop  was  made  for. 

Hughes:  This  confluence  of  disciplines,  that  we  talked  about,  has  so  many 
ramifications.  One  of  them  is  bringing  in  disciplines  that  don't 
have  a  long  background  in  working  safely  with  pathogens. 


1  For  example,  see  Edwin  Lennette,  Pioneer  of  Diagnostic  Virology  with 
the  California  Department  of  Public  Health,  Regional  Oral  History  Office, 
The  Bancroft  Library,  University  of  California  at  Berkeley,  1988. 


79 


Berg:    That's  right.   If  biochemists  broke  and  spilled  an  important 

experiment,  they'd  suck  it  up  without  thinking,  even  if  it  was 
something  dangerous.   Their  goal  is  to  save  the  experiment. 


Achievements  of  the  Research  Moratorium 


Berg:     Well,  if  you  were  working  with  really  hazardous  things,  you'd  say 
that  kind  of  behavior  is  unacceptable.   But  if  you  were  working  on 
problems  at  open  benches,  you  don't  know  how  such  people  think 
about  risks.   What  does  it  take  to  do  so?  What  kind  of  ethics  or 
whatever  does  it  take  for  somebody  to  say,  wait  a  minute,  you 
don't  want  to  do  something  so  stupid  that  it's  dangerous  to 
yourself,  to  your  family,  to  your  co-workers.   It  doesn't  take  any 
higher  calling  for  somebody  to  say,  "Hey,  wait  a  minute,  we  ought 
to  think  a  little  bit  about  what  we're  doing  to  see  whether  it's 
safe." 

Hughes:   And  that's  what  Asilomar  II  and  that  complex  period  of  history 
did? 

Berg:     That's  right.   Today,  there's  no  question  that  it  impeded  some 

research.   But  my  argument  is  that  impedance  actually  benefitted 
the  research  in  the  long  run.   For  one,  I  think  we  didn't  generate 
the  kind  of  public  opposition  that  could  have  easily  stopped  the 
research.   Because  we  took  the  initiative,  and  brought  attention 
to  the  problem,  and  tried  to  deal  with  it,  people  accepted  that  we 
were  conscientious,  well-meaning,  and  responsible. 

Second  of  all,  I  don't  know  how  much  work  on  cloning  human 
genes  could  have  been  done  right  from  the  beginning.   We  didn't 
know  how  to  walk,  no  less  run.   We  barely  could  crawl.   When  the 
Asilomar  constraints  said  you  can  do  this  under  these  conditions 
and  those  conditions,  people  learned  how  to  do  the  cloning  under 
safe  conditions.   People  learned  to  adapt  so  that  now  nobody  ever 
used  mouth  pipettes;  people  worked  in  hoods;  people  had  to  take 
care.   And  before  you  knew  it,  it  was  possible.   The  technology 
had  improved.   People's  approach  to  things  had  changed.   You  could 
now  go  after  human  genes.   So,  I  don't  think  that  we  lost  a  lot  by 
saying  you  couldn't  clone  human  DNA  for  the  first  year  or  two. 
But  it  certainly  prevented  people,  or  called  attention  to  the  fact 
that  you  had  to  think  about  what  you  were  going  to  do  before,  not 
after.   And  you  had  to  think  about,  was  it  sensible?  Was  there 
any  kind  of  a  risk?  Could  I  learn  what  I  want  to  learn  another 
way? 


80 

Hughes:   Well,  there  were  some  examples  of  things  that  had  gone  wrong, 

particularly  in  Britain.   I  understand  that  they  colored  the  Ashby 
group's  considerations.  What  was  it?  Smallpox? 

Berg:    Yes. 

Hughes:   There  had  been  some  recent  deaths  in  a  British  laboratory.   As  you 
say,  if  the  research  momentum  had  continued  unabated— 

Berg:    Somebody  would  have  done  a  dumb  experiment. 

But  you  also  have  to  remember  that  there  were  scientists, 
who  had  a  very  different  perspective—Science  for  the  People  kind 
of  thing.   They  were  primarily  left-leaning  people.   The  Vietnam 
War  spawned  a  whole  lot  of  people  to  be  very  suspicious,  not 
accepting.   There  were  claims  being  made  about  certain  genes 
predisposing  to  criminality.  And  this  group  in  Boston  [Science 
for  the  People]  reacted  to  that. 

Had  we  not  called  attention  to  what  we  were  doing 
[recombinant  DNA  research],  they  would  have.   I  believe  they  would 
have  said  to  the  public,  "Look  at  what  these  guys  are  doing;  this 
is  really  dangerous  stuff.   It's  the  first  step  to  genetic 
manipulations  of  man,"  and  blah,  blah,  blah,  which  is  what  they 
tried  to  do.   But  given  that  we  had  raised  the  safety  issue  first 
and  tried  to  deal  with  it,  I  think  their  message  was  blunted.   And 
while  they  were  harassing  us  much. of  the  time  and  were  successful 
in  some  places,  Cambridge  and  so  on,  in  the  end  they  didn't  win. 
And  the  science  moved  forward. 

So  as  I  look  back  on  the  period,  even  though  we  were  wrong- - 
wrong  is  probably  not  the  right  word;  certainly  our  assessment  of 
the  potential  risk  was  incorrect—by  calling  attention  to  it,  I 
think  the  whole  thing  was  better  off  in  the  long  term.   The 
science  that  has  come  out  of  it  has  just  been  absolutely  mind- 
boggling.   And  so  that's  what  in  the  end  will  justify  it. 


Transduction 

[Interview  3:  September  30,  1997] 

Lysogeny  and  Transduction 


Hughes:   Dr.  Berg,  my  goal  today  is  not  to  talk  exhaustively  about  the 
biohazard  issue,  which  has  been  well-covered  in  the  historical 


81 


literature.1  Instead,  I  want  to  focus  on  the  science.   What  were 
the  technologies  that  made  the  recombinant  DNA  work  possible? 

Berg:    Well,  there  were  really  two  lines  of  investigation  that  were 
important.   I  think  I  ended  my  last  interview  talking  about 
lysogeny.   Lysogeny  is  a  phenomenon  in  which  viruses  that  infect 
bacteria  can  integrate  their  chromosome  into  the  chromosome  of  the 
infected  cell.  And  then  they  are  maintained  as  if  they  were  a 
normal  part  of  that  organism's  chromosome.   They're  replicated 
each  cycle.   The  cells  are  perfectly  healthy.   They  reveal  some 
new  properties  as  a  result  of  having  acquired  the  virus 
information.   But  with  low  frequency,  this  mutually  acceptable 
state  breaks  down  and  the  viral  chromosome  pops  out  of  the 
cellular  chromosome  and  replicates,  killing  the  cells  and 
producing  the  virus.   That  state  is  referred  to  as  lysogenic, 
meaning  that  the  cells,  while  they're  perfectly  normal  and  grow 
perfectly  well,  enter  a  state  after  the  virus  is  activated,  and 
the  virus  kills  the  cells  by  lysing  them. 

Now,  if  you  ask,  what  is  the  virus  that  comes  out?   Is  it 
exactly  the  same  as  the  virus  that  went  in?   And,  99.999  percent 
of  the  viruses  that  come  out  are  exactly  the  same  as  those  that 
infected  the  cells  originally.   That  is,  they  are  excised  from  the 
cellular  chromosome  perfectly  accurately.   But,  occasionally,  they 
actually  come  out  with  some  of  the  adjacent  cellular  genes.   And 
so,  the  virus  that  comes  out  now  is  different  than  what  went  in. 
It  has  lost  something  of  its  own  chromosome  and  picked  up  some  of 
the  cellular  chromosome.   Such  viruses,  when  they  infect  the  next 
cell,  will  be  able  to  transfer  genes  from  the  original  cell  they 
were  in  to  the  new  cell  they  infect.   That  process,  called 
transduction,  was  discovered  many  years  ago  by  Norton  Zinder  and 
Joshua  Lederberg.   So  depending  upon  where  the  virus  integrates, 
it's  able  to  pick  up  genetic  information  of  the  cell  closely 
linked  to  it. 


Other  Forms  of  Transduction 


Berg:    There's  a  second  phenomenon  of  transduction  in  which  another 

virus,  a  different  one,  goes  in  and  infects  the  cells  and  kills 
the  cells.   And,  in  the  act  of  killing  the  cells,  it  literally 
pulverizes  the  cellular  chromosome  into  bits  and  pieces.   In  the 


1  See,  for  example:  Susan  Wright,  Molecular  Politics:  Developing 
American  and  British  Regulatory  Policy  for  Genetic  Engineering,  1972-1982, 
Chicago:  University  of  Chicago  Press,  1994. 


82 


packaging  event,  which  leads  to  the  production  of  new  virus 
particles,  these  bits  and  pieces  of  cellular  chromosome  are 
inadvertently  packaged  into  the  viral  particle.   As  a  consequence, 
a  population  of  viruses  is  produced  which  carries  different  pieces 
of  the  cellular  chromosome  as  well  as  their  own.   If  you  looked  at 
these  virus  particles,  you  couldn't  tell  the  difference  as  to 
whether  they  had  their  own  chromosome  or  whether  they  carry  pieces 
of  the  cellular  chromosome.   They  infect  cells;  they  bring  in  this 
new  genetic  information.   And  this  genetic  information  can  then 
replace  what  is  present  in  the  cell.   And  so,  you  now  get  a  new 
genetic  property.   This  process  is  referred  to  as  transduction. 

So,  there  are  two  kinds  of  transducing  viruses.   One  of  them 
is  what  we  call  lysogenic,  the  virus  goes  in,  integrates,  stays 
there  for  however  long,  spontaneously  pops  out,  most  of  the  time 
accurately.   Occasionally,  it  picks  up  cellular  genes  alongside 
it.   And  if  you  know  where  the  virus  goes,  you  can  actually  then 
transfer  genes  that  are  adjacent  to  the  site  of  integration. 

Hughes:   This  was  all  well  known? 
Berg:     All  well  known. 
Hughes:   By  when  would  you  say? 

Berg:     I  was  doing  this  in  conjunction  with  Charles  Yanofsky,  who's  a 
geneticist.   This  is  the  technique  of  modifying  bacterial  cells 
and  introducing  new  kinds  of  genes  in  them.   For  that  purpose  we 
used  bacteriophage  lambda  or  phi  80.   Those  are  two  kinds  of 
phages.   They  each  integrate  in  different  places  in  the  host 
chromosome.   We  used  the  phi  80  phage  because  it  often  could  carry 
the  genes  that  controlled  tryptophan  synthesis,  and  we  were 
studying  tryptophan  mutants.   We  could  transduce  genes  from  one 
cell  into  another  and  create  cell  lines  that  had  useful  genetic 
properties  for  our  experiments. 

The  lambda  phage  goes  into  a  different  part  of  the  E.  coli 
chromosome,  and  it  carries  different  sets  of  genes  when  it  comes 
out.   The  P,  phage  is  different  because  it  picks  up  random  pieces 
of  the  cellular  DNA.   That's  also  very  useful  because  in  any 
population,  a  virus  that  comes  out  of  an  infection  will  carry 
genes  that  you're  interested  in  studying.   Using  Pj  you  can 
transfer  any  genes. 

So,  the  important  conceptual  point  is,  there  was  a  way  to 
transfer  genes  from  one  cell  to  another,  and  it  had  proven  to  be 
extremely  useful  and  powerful  in  setting  up  experimental  systems 
for  genetic  studies. 


83 


Hughes:   Is  this  a  potential  evolutionary  mechanism? 

Berg:    Usually  these  viruses  are  quite  specific  for  the  cells  they 

infect.   So,  it  isn't  that  they  spread  their  genes  throughout  a 
population.   But  certainly  within  a  certain  group  of  organisms, 
genes  are  flowing  back  and  forth.   No  question  about  that.   I 
suppose  if  you  put  a  selection  on,  that  is  conditions  which  favor 
the  growth  of  cells  that  contain  one  set  of  genes,  those  will  have 
preferential  growth,  and  the  others  will  die  out. 

Hughes:   So,  there  was  a  natural  mechanism  that  you  took  advantage  of. 
Berg:     That's  right. 

Hughes:   What  couldn't  you  do  with  it  experimentally  that  you  hoped  to  do? 
Why  devise  an  artificial  laboratory  mechanism  for  transferring 
genes? 

Berg:     Because  you  could,  in  fact,  use  conditions  that  selected  for  just 
the  kinds  of  organisms  you  want.   By  doing  the  transduction  in 
several  ways,  we  were  able  to  adapt  that  natural  phenomenon  to  our 
advantage.   Now,  this  was  true  of  bacteria  and  bacteriophages.   1 
would  attribute  a  very  large  part  of  the  burst  of  genetic 
knowledge  during  the  1960s  to  the  astute  use  of  these 
bacteriophages.   That  certainly  helped.  Another  thing  is,  once 
these  viruses  come  out,  you  can  isolate  the  ones  that  carry  these 
cellular  genes,  and  then  you  begin  to  analyze  the  sequence. 

In  one  case,  it  was  possible  to  sequence  a  large  part  of  one 
region  of  the  bacterial  chromosome  because  it  was  now  highly 
enriched  in  the  bacteriophage  DNA.   After  all,  the  viral 
chromosome  was  tiny  compared  to  the  cellular  chromosome,  so  you 
get  an  enormous  enrichment.   If  you  harvest  the  virus,  you've  now 
looked  at  a  small  portion  of  the  cellular  chromosome  and  can 
analyze  that.   And  that  was  done. 


Transduction  of  Mammalian  Cells 


Hughes:   You  said  that  you  had  a  natural  system  that  you  were  taking 

advantage  of.   What  I  see  coming  next,  with  recombinant  DNA  work, 
is  that  you  created  constructs  for  scientific  purposes.  No  longer 
were  you  just  taking  advantage  of  what  nature  had  already 
provided. 

Berg:     That's  right.   But  the  important  thing  is  to  realize  that  in 

mammalian  cells,  there  was  no  such  natural  system.   No  viruses 


were  known  that  could  transduce  genes  to  mammalian  cells.   Well, 
let  me  put  it  this  way.   When  I  went  to  the  Salk  Institute  for  a 
sabbatical,  it  was  to  learn  about  how  to  work  with  mammalian 
cells.   The  motivation  formed  from  the  fact  that  viruses  had  been 
so  influential  and  important  for  the  development  of  the  molecular 
biology  of  microorganisms  that  maybe  studying  the  interaction  of 
animal  viruses  with  animal  cells  would  give  us  the  same  kind  of 
insights. 

The  polyoma  virus  I  studied  has  a  very  small  chromosome, 
only  five  genes.   So,  if  you  wanted  to  study  how  a  cell  expresses 
its  genes,  instead  of  looking  at  how  it  expresses  its  own  genes, 
why  not  look  at  how  it  expresses  the  virus  genes  once  they  enter 
the  cell—a  much  easier  system.   So,  when  I  went  to  Salk,  it  was 
to  begin  to  understand  something  about  how  these  tumor  viruses 
multiply  in  a  mammalian  cell. 

We  chose  the  tumor  virus  because  it  too  mimics,  to  some 
extent,  lysogenic  viruses.   It  can  integrate  into  the  cellular 
chromosome.   And  we  thought,  well,  perhaps  under  certain 
conditions  it  could  come  out  and  carry  with  it  genes  that  are 
adjacent  to  where  it  had  integrated.   During  that  year,  we  had  a 
lot  of  success  studying  some  interesting  features  about  polyoma 
infection  of  mouse  cells. 

One  of  the  things  that  others  discovered  was  that  there  were 
virus  particles  coming  out  carrying  cellular  DNA.   That  looked 
like  the  P!  phage .   So  the  question  was,  could  we  in  fact  use  such 
viruses  in  the  same  way  that  the  bacterial  people  had  used  P,  as  a 
way  of  transporting  genes  into  mammalian  cells?  The  idea  was  to 
grow  the  viruses  in  one  kind  of  cell,  then  take  the  population  of 
viruses  that  come  out  and  infect  another  population  of  cells,  and 
ask  whether  interesting  genes  were  transferred  from  one  to  the 
other. 

Hughes:   How  at  that  time  did  you  distinguish  viral  from  mammalian  DNA? 

Berg:    We  could  label  the  DNA  of  the  virus,  and  we  could  follow  its 

transactions,  if  you  will,  once  it  entered  the  cell.   Two,  once  we 
have  the  DNA  of  the  virus,  we  can  use  that  DNA  as  a  probe.   When 
the  virus  enters  the  cell,  it's  transcribed  and  makes  messenger 
RNAs.   If  we  had  the  pure  DNA,  we  could  use  the  DNA  to  detect 
those  messengers.   But  since  isolated  genes  from  mammalian  cells 
were  not  available,  there  was  no  way  to  follow  the  transcription 
or  expression  of  mammalian  genes. 

Remember  that  the  mammalian  genome  is  larger  and  more 
complex  than  the  bacterial  genome,  and  the  amount  of  DNA  is  a 
thousand  times  greater  than  is  present  in  a  bacterium.   So,  the 


85 


idea  of  being  able  to  follow  the  expression  of  mammalian  genes  was 
very  complicated.   There  were  a  lot  of  puzzles.   For  example,  when 
people  tried  to  look  at  something  that  was  supposed  to  be  the 
equivalent  of  messenger  RNA,  they  found  an  enormously  complex 
mixture  of  RNAs  of  varying  sizes;  most  of  it  never  ended  up  as 
messenger  RNA.   That  was  a  real  puzzle.   In  bacteria,  you  could 
measure  transcription.  An  RNA  molecule  is  colinear  with  the 
segment  of  DNA  from  which  it  is  transcribed.   But  in  mammalian 
cells  there  was  this  mess,  a  real  mess,  98  percent  of  which  never 
makes  it  into  the  cytoplasm. 

Hughes:   You  didn't  find  that  intimidating? 

Berg:    No.   Some  people  did.   Kornberg  tried  to  persuade  me  not  to  get 

involved  in  such  a  messy  system;  "You're  wasting  your  talent,"  he 
told  me.   He  was  really  very,  very  critical  of  the  decision  to 
enter  this  field.   However,  I  was  convinced  that  we  were  beginning 
to  know  a  lot  about  the  bacterial  genetic  system--how  it's 
expressed,  how  it's  replicated,  and  so  on.   There  were  elaborate 
theories  of  regulation,  of  the  messenger  RNA  concept.   One  had  to 
stop  and  ask  oneself,  Is  all  of  this  unique  to  the  bacterium?   Is 
this  the  way  things  happen  in  higher  organisms?  Well,  I  wanted  to 
learn  if  this  whole  system  of  messenger  RNAs,  transcription, 
operators,  repressers,  polymerases,  and  so  on,  existed  in 
mammalian  cells.   How  do  you  begin  to  study  mammalian  cells?   That 
is  intimidating.   The  virus  was  the  key.   The  virus  was  the  way  to 
get  into  that  system  and  simplify  it. 

Hughes:   So,  the  virus  approach  made  it  acceptable  to  you  to  do  this  very 
complicated,  risky  research? 

Berg:     Right.   In  fact,  it  turned  out  to  be  correct;  the  virus  was  the 
simple  way  to  be  able  to  look  at  it.   And  people  just  went  to 
different  viruses.   We  began  to  work  with  SV40;  other  people 
worked  with  polyoma,  adenovirus .   But  the  virus  was  the  entree 
into  studying  how  mammalian  cells  deal  with  a  piece  of  DNA.   Use  a 
small  piece  of  DNA  [the  virus]  so  that  you  can  distinguish  it  from 
the  cellular  DNA. 

During  the  year  at  the  Salk  it  was  discovered  that  viruses 
coming  out  of  the  infected  cells  seemed  to  be  carrying  some 
mammalian  DNA  sequences.  We  thought,  perhaps  is  this  the  analog 
of  the  P]  transduction  system?   If  you  make  the  calculation,  you 
soon  realize  that  SV40  can  only  package  about  5,000  base  pairs  of 
DNA,  whereas  the  bacteriophages  that  we  were  using  before  could 
package  50,000  b.p. 


86 


Hughes:   What  are  the  limitations? 

Berg:    The  limitations  are  the  size  of  the  viral  capsid  or  shell  capsid. 
The  capsid  has  to  be  built,  and  only  so  much  DNA  can  be  stuffed  in 
it.   So,  5,000  base  pairs  of  DNA  go  in.   Now,  if  you  ask,  given 
the  size  of  the  mammalian  genome,  3  billion  base  pairs,  a  5,000 
base  pair  segment  of  it  is  a  very,  very  small  fraction  of  the 
total.   So,  the  question  is,  any  one  virus  particle  could  only 
contain  an  inf initesimally  small  amount  of  the  cellular 
chromosome.   If  you  were  looking  for  a  particular  gene,  it  would 
be  very  rare.  And  if  it  was  very  rare,  you  would  have  to  have  an 
incredibly  powerful  way  of  detecting  its  transfer  and  ultimate 
function.   And  so  we  quickly  realized  it  was  hopeless  to  use  this 
as  an  analog  of  P,.   Of  course,  we  knew  nothing  about  the 
likelihood  of  being  able  to  incorporate  a  whole  gene.   We  didn't 
know  the  size  of  mammalian  genes.   Now  we  know  it  would  have  been 
impossible. 

At  that  time  we  thought  mammalian  genes  were  about  the  same 
size  as  bacterial  genes.   You  might  have  been  able  to  package  them 
but  it  would  have  been  so  rare  that  only  maybe  one  in  a  billion 
particles  would  actually  contain  the  gene  of  interest  in 
functional  form.   And  then  you  would  have  the  job  of  finding  a 
needle  in  the  haystack.   You'd  have  to  have  an  enormous  population 
of  mammalian  cells  infected  by  an  enormous  population  of  virus 
particles  in  order  to  detect  the  transfer  of  one  gene. 

Hughes:   So,  detection  is  really  key,  isn't  it? 

Berg:     Logistics,  detection,  and  the  packaging  limitation  in  the  virus 
were  all  things  that  precluded  what  we  had  in  mind,  namely  that 
you  could  transfer  genes  using  SVAO  viruses  as  vectors.   If  you 
had  viruses  that  could  carry  much  more  DNA,  you  might  have  had  a 
chance. 


More  on  Recombinant  DNA  Science 


Construction  of  Recombinant  DNA  Molecules  to  Study  the 
Mammalian  Cell 


Berg:    So  we  said,  okay,  that's  not  going  to  work.   Can  we  construct  DNA 
molecules  which  in  fact  use  the  virus  chromosome?  Let's  just 
attach  foreign  pieces  of  DNA,  any  genes  we  might  ultimately  want, 
to  the  viral  DNA.   At  the  time,  you  have  to  remember,  nobody  had 
isolated  any  genes.   I  believed  that  that  would  eventually  be 


87 


achieved.   People  would  find  ways.   One  hint  that  this  was  likely 
was  that  certain  classes  of  genes  have  physical  properties  which 
allowed  them  to  be  separated  from  the  bulk  of  cellular  DNA. 

Don  Brown,  at  the  Carnegie  Labs  in  Baltimore,  was  able  to 
isolate  pure  ribosomal  RNA  genes,  because  they  have  an  unusual 
buoyant  density.   That  is,  if  you  put  them  in  a  centrifuge  in  a 
gradient  of  salt  concentration,  the  ribosomal  DNA  genes  separate 
from  the  rest  of  the  DNA.  And,  you  can  purify  them  that  way. 
Nobody  had  isolated  any  other  genes,  but  it  wasn't  too  far-fetched 
that  there  would  be  a  way  to  isolate  individual  genes  sometime  in 
the  future.   So,  on  that  premise  we  said,  "We  need  to  develop  a 
method  for  attaching  any  piece  of  DNA  to  SVAO  DNA." 

Hughes:   Was  your  premise  based  on  the  fact  that  you  had  a  technique  which 
could  be  developed  further,  or  was  it  an  expression  of  optimism 
that  science  will  find  the  answer? 

Berg:     There  was  a  certain  amount  of  optimism,  faith,  however  you  want  to 
refer  to  it.   But,  in  this  particular  case,  I  think  what  we  were 
asking  was  a  question  that  was  wholly  within  our  own  domain. 
Could  we  devise  a  way  to  attach  any  piece  of  DNA  to  SV40  so  that, 
if  cells  were  exposed  to  this  recombinant  DNA,  they  would  take  it 
up.  If  so,  there  would  be  a  way  of  introducing  this  DNA  into  a 
mammalian  cell. 

Now,  this  piece  of  DNA  could  be  nondescript,  that  is,  from 
any  source.   From  a  technical  point  of  view,  all  we  needed  to  do 
was  to  learn  how  to  attach  two  pieces  of  DNA.   The  faith  and 
optimism  was  that  down  the  line  people  were  going  to  find  ways  to 
prepare  genes  that  would  be  interesting  to  put  into  cells. 

So,  for  our  purposes,  we  just  started  with  a  piece  of 
bacterial  DNA  which  had  three  genes  whose  properties  we  knew 
something  about.   And  if  these  were  attached  to  SV40  introduced 
into  mammalian  cells,  we  could  test  if  bacterial  genes  could 
function.   That  was  a  totally  unanswered  question.   As  we  know 
today,  they  would  not  have  been  functional.   But,  we  didn't  know 
that  then.   So,  the  question  was,  if  you  could  in  fact  take  genes 
from  other  organisms  and  attach  them  to  SV40  and  piggyback  them 
into  the  mammalian  cell,  would  they  function?   And  if  they 
functioned,  could  you  learn  something  interesting  about  the 
mammalian  cell? 


88 


Bacteriophage  with  Cohesive  Ends 


Berg:    The  question  was,  How  do  you  attach  two  pieces  of  DNA?  So,  now 
comes  the  second  bit  of  information  that  was  standard  lore.   You 
asked  me  about  the  precursors.   People  had  been  studying  lysogenic 
bacteriophages,  e.g.  bacteriophage  lambda  and  phi  80.   The  DNA  in 
phage  lambda  is  about  50,000  base  pairs  long,  and  it  exists  in  the 
virus  particle  as  a  linear,  double-stranded  DNA.   What  was 
discovered  is  that  these  viruses  had  unusual  ends,  sticky  ends. 
They  had  protrusions  of  single  strands  from  each  end.   These 
single-strand  ends  are  complementary  to  each  other  so  that,  even 
though  the  DNA  from  the  bacteriophage  is  clearly  linear,  the  DNA 
circularizes  when  subjected  to  very  simple  conditions.   They 
circularize,  and  they  circularize  because  the  two  single  strands 
come  together  and  form  double  strands.   These  linear  molecules  can 
also  form  long  chains,  because  they  attach  end  to  end  through  the 
cohesive  ends.   And  that's  the  origin  of  the  term  sticky  ends  or 
cohesive  ends. 

Hughes:   That  was  work  done  here  at  Stanford? 

Berg:    Well,  some  of  that  work  was  done  here  by  Dale  Kaiser,  who  was  our 
principal  person  in  bacteriophage  and  one  of  the  major  workers  in 
the  field  of  lysogeny.  Alfred  Hershey,  who  was  then  at  Cold 
Spring  Harbor  Laboratory,  was  the  initial  discoverer  of  cohesive 
ends.   It  was  learned  later  that  this  class  of  lysogenic  phages 
had  cohesive  ends.   But  the  ends  are  all  different.   So,  if  you 
take  lambda  phage  and  phi  80,  they  will  not  join  to  each  other, 
because  their  ends  are  different.   Sticky  ends  are  important  in 
the  reproduction  of  the  virus.   The  virus  goes  in  as  a  linear 
molecule,  and  once  inside  the  cell,  it  circularizes  and  the  nicks 
between  the  ends  get  closed.   And  then  it  functions  as  a  circular 
DNA  molecule. 


Creating  Artificial  Cohesive  Ends 


Berg:    So,  the  concept  of  sticky  ends  already  existed.   If  you  want  to 

join  two  different  molecules  together,  it  doesn't  take  a  genius  to 
figure  out  that  if  you  can  create  artificial  ends  that  are 
complementary  to  each  other  the  two  DNA  molecules  will  come 
together.   Right?  No  big  deal. 

So,  if  you  put  tails  of  A  on  one  piece  of  DNA  and  tails  of  T 
on  SV40  DNA,  and  mix  them,  the  A's  and  T's  will  form  double 


89 


helices,  and  the  two  molecules  will  come  together.   Each  DNA 
cannot  join  to  itself,  only  to  one  with  complementary  tails.   We 
could  have  used  G's  and  C's,  but  A's  and  T's  were  easier  to  add. 

We  already  knew  how  to  add  tails  onto  DNA  molecules  because 
there  is  an  enzyme  that  had  been  described  which  is  present  in 
calf  thymus;  it  has  an  interesting  physiological  function,  but 
that  was  not  known  at  the  time.   It  is  a  DNA  polymerase,  but  a 
"dumb"  DNA  polymerase.   It  doesn't  need  a  template.   If  you  give 
it  any  one  of  the  four  deoxynucleoside  triphosphates,  it  will  add 
the  nucleotide  on  to  the  end  of  the  DNA  molecule,  producing  long 
chains  of  the  same  nucleotide.   So,  if  DNA  molecule  A  is  mixed 
with  deoxyATP  and  this  enzyme,  long  polymers  of  A's  are  added  onto 
the  two  3-prime  ends  of  this  DNA.   And  if  you  do  it  with  deoxyTTP, 
long  chains  of  T's  are  added.   By  regulating  the  time  of  the 
reaction,  you  can  add,  on  average,  about  100  A's  or  T's  onto  each 
end. 

Hughes:   Who  worked  out  that  procedure? 

Berg:     The  enzyme  was  found  by  Fred  Bollum  and  it  is  called  nucleotidyl 
terminal  transf erase.   This  enzyme  cannot  make  DNA  molecules  de 
novo  from  deoxytriphosphates ;  it  always  needs  a  primer  end.   And, 
we  knew  that.   Therefore,  if  we  took  a  DNA  molecule,  its  3-prime 
ends  served  as  primers  to  polymerize  onto  the  ends.   Then  adding 
T's  to  the  ends  of  a  second  DNA,  they  would  come  together  to  form 
circles . 


Enzymatic  Sealing  of  DNA  Circles 


Berg:     Now,  the  dA  and  dT  tails  are  uneven  because  there's  no  way  to  get 
them  precisely  the  same  length.   When  the  two  DNAs  are  joined, 
there  are  gaps  at  the  join.   Kornberg's  DNA  polymerase  was  the 
perfect  thing  to  fill  in  gaps;  we  knew  that.   We  knew  that  once  we 
made  circles  they  would  have  gaps,  but  these  could  be  filled  with 
DNA  polymerase.   You  just  add  all  four  deoxytriphosphates,  and  the 
enzyme  fills  in  the  gaps.  And  if  you  add  the  enzyme  DNA  ligase, 
which  had  also  been  co-discovered  at  Stanford  by  Bob  Lehman,  the 
ends  become  covalently  joined.   So  by  taking  these  two  separately 
prepared  DNA  molecules,  mixing  them,  waiting  a  few  minutes  until 
they  anneal,  adding  DNA  polymerase,  four  deoxytriphosphates,  and 
DNA  ligase,  then  you  have  a  recombinant  DNA. 

The  recombinant  DNA  we  made  involved  joining  SV40  together 
with  the  piece  of  DNA  that  contained  three  bacterial  genes.   The 


90 


reason  we  could  use  these  three  bacterial  genes  is  because  they 
had  been  picked  up  by  lambda  by  this  kind  of  inexact  excision. 


Biochemistry  Department  Contributions  ft 


Hughes:   You  had  all  the  ingredients  to  perform  this  experiment  right  there 
in  the  biochemistry  department? 

Berg:    In  a  refrigerator  which  I  had  complete  access  to.   That's  a 

terribly  important  point.   I've  made  that  point  many,  many  times 
over.   It's  a  reflection  of  the  kind  of  atmosphere  that  we  had  in 
our  department,  the  kind  of  relationships  which  we  had  with  each 
other,  which  was  completely  giving  and  open.   It  wasn't 
competitive.   It  wasn't  secretive.   I  had  access  not  only  to  all 
of  the  information  that  would  ultimately  be  needed  to  do  this 
experiment,  but  in  addition,  the  materials  were  accessible.   I 
didn't  have  to  stop  to  make  these  enzymes.   We  made  some.   But  the 
point  is  they  were  all  there  and  the  expertise  for  how  to  use 
them. 

Hughes:   Very  important. 

Berg:    Very  important.   And  so,  the  actual  accomplishment  was  quite 
straightforward. 


Replication  and  Expression  in  the  Host  Cell 


Berg:     Now,  we  intended  to  introduce  these  recombinant  DNA  molecules  into 
mammalian  cells.   And  the  question  was,  would  the  bacterial  genes 
function  that  were  introduced  along  with  the  SV40  DNA?  Before  we 
got  the  chance  to  do  that  experiment,  it  was  recognized  that  the 
piece  of  bacterial  DNA  we  had  used  also  was  capable  of  replicating 
in  bacteria.   So  in  principle,  this  recombinant  DNA  could  also  be 
put  into  bacteria  to  determine  if  the  recombinant  DNA  would 
replicate.   And  would  the  SV40  genes  be  expressed  in  bacteria? 

Hughes:   Was  that  a  question  that  you  asked  at  the  outset? 

Berg:  No,  that  was  not  a  question  that  was  in  our  plans.  Some  people, 
particularly  Stan  Cohen,  thought  that  was  what  we  were  aiming  to 
do. 


91 
Concern  about  Potential  Biohazards 

Janet  Mertz  at  the  Tumor  Virus  Course 


Berg:    But  in  point  of  fact,  what  happened,  and  this  is  part  of  the 

recombinant  DNA  story,  is  that  one  of  my  students,  Janet  Mertz, 
went  off  to  Cold  Spring  Harbor  during  the  summer  of  1971  to  take  a 
tumor  virus  course.  As  a  student  in  the  course,  she  was  asked  to 
give  a  seminar  on  what  research  she  was  doing,  and  she  described 
what  we  were  doing.   That  aroused  enormous  anxiety. 

Bob  Pollock,  one  of  the  instructors  in  the  course,  got  all 
exercised  about  the  possibility  that  we  would  be  putting  genes 
from  a  tumor  virus  into  a  bacterium  which  normally  inhabits 
humans.   Bob  called  me  from  Cold  Spring  Harbor  asking,  "Why  are 
you  doing  this  crazy  experiment?"  He  was  referring  to  the 
possibility  of  introducing  recombinant  DNA  into  £.  coli.   That 
wasn't  in  our  mind  at  all.   During  all  of  the  furor,  concern  was 
raised  that  just  making  these  molecules  in  the  lab  might  allow 
them  to  come  in  contact  with  bacteria,  and  these  would  spread  a 
plague  of  cancer. 


Berg's  and  Lederberg's  Reactions 


Hughes:   According  to  the  accounts,  Janet  Mertz  called  you  from  Cold  Spring 
Harbor.   Can  you  remember  your  first  reaction? 

Berg:     My  first  reaction  was,  this  is  stupid;  this  is  bunk.   I  thought  it 
was  outrageous.   Then,  as  we  talked  a  bit,  1  kept  trying  to  think 
of  ways  to  sort  of  fend  off,  or  even  respond  to,  the  concerns  by 
saying  this,  that,  or  the  other  thing.   By  that  time,  it  was  clear 
that  I  was  being  forced  to  consider  what  might  happen  in  an 
experiment  that  we  hadn't  even  thought  about  doing.   We  kept 
talking  about  different  ways  to  prevent  what  was  of  concern.   But, 
given  how  paranoid  the  concerns,  I  began  to  ask  myself  if  there 
was  a  small  possibility  of  risk.  And  if  there  is,  do  I  want  to  do 
the  experiment? 

I  remember  going  to  talk  to  Josh  Lederberg  because  he  had 
already  been  talking  about  making  germ  warfare  weapons,  using  the 
new  molecular  genetics,  as  we  do  with  the  bacteria.   Nothing  had 
been  talked  about  using  mammalian  cells  or  animal  viruses  or 
anything.   And  Josh,  interestingly  enough,  was  very,  very 
conservative.   I  mean,  he  was  not  about  to  say,  "Go  ahead  and  do 


92 


this  experiment."  Instead  he  said,  "There's  certainly  the 
possibility,  and  you  have  to  take  responsibility."  At  that  point 
I  stepped  back  and  asked,  Do  I  want  to  go  ahead  and  do  experiments 
which  could  have  catastrophic  consequences,  no  matter  how  slim  the 
likelihood? 

Hughes:   Yes,  and,  as  you  said  last  time,  you  had  already  had  concerns  in 
this  general  area.   You  were  the  convener  of  Asilomar  I. 

Berg:    And  Pollock  was  one  of  the  key  people  in  that  too;  in  fact  we  had 
been  its  co-organizers. 

Hughes:   Hadn't  Lederberg  been  involved  with  advising  on  space  probes  and 
the  potential  danger  of  bringing  foreign  microorganisms  to  this 
planet? 

Berg:    Yes,  absolutely,  it  was  in  the  sixties. 

Although  my  first  reaction  to  Pollock's  call  was  outrage,  I 
had  many  conversations  with  him  by  phone.   Since  I  knew  Bob  quite 
well  and  thought  we  had  a  very  good  relationship,  I  couldn't  just 
fluff  off  what  he  was  concerned  about.   Then  I  began  to  talk  to 
still  other  people,  a  fellow  named  Ted  Friedman  who  had  also  been 
influential  in  organizing  the  first  Asilomar  conference.   Ted  had 
been  a  strong  advocate  of  ethics  in  science.   I  was  barraged  by 
hypotheticals,  and  they  were  all  hypotheticals .   There  was 
certainly  no  evidence  that  if  a  bacterium  carrying  genes  that 
produce  cancer  in  man  got  into  your  gut  that  this  would  inevitably 
cause  cancer.   There  was  certainly  no  indication  that  this  was  the 
case . 


Putting  the  SV40  Experiment  on  Hold 


Berg:     I  guess  in  the  end  I  finally  said,  well,  let's  step  back  and  ask, 
is  there  another  way  to  answer  the  question  for  which  this 
experiment  was  designed?  The  experiment  was  designed  to  ask,  can 
we  get  genes  into  mammalian  cells,  and  if  we  do,  do  they  function? 
The  question  was,  could  you  set  up  a  different  kind  of 
experimental  system  which  would  not  create  a  viable  organism  that 
could  be  spread.   And  so,  we  put  the  experiment  on  hold. 

I  suspect  we  would  not  have  put  the  experiment  on  hold  had 
Dave  Jackson  not  taken  a  job  and  gone  off  to  Michigan.   The  fact 
is  that  he  had  accepted  this  job  and  was  due  to  go  to  Michigan 
even  before  he  finished  the  recombination  experiment.   And  it  was 
only  because  I  called  his  new  chief  and  said  I  was  not  going  to 


93 


Hughes : 


let  him  go;  we're  going  to  chain  him  to  the  bench.   He  had  to 
finish  the  experiment  before  he  left  because  I  knew  that  once  he 
left,  the  chances  of  him  continuing  it  in  a  new  lab  were  small. 
And  so,  he  stayed.   Once  we  got  the  paper  written,  he  left.1 

The  next  step  would  have  been  to  introduce  this  DNA  into 

mammalian  cells  and  do  the  experiment.  And  that  was  trivial.   I 

mean,  it  would  have  been  done.   But,  with  all  of  the  furor  that 
got  raised,  we  just  decided,  Let's  hold  off. 

So,  there  were  two  things  that  were  stopped.   There  was  the 
transfer  of  the  recombinant  construct  into  mammalian  cells  and 
also  into  bacterial  cells. 


Berg:     That's  right.   The  concern  was  primarily  about  the  bacterial 
cells.   Nobody  raised  any  serious  objection  to  putting  the 
construct  into  mammalian  cells.   They  raised  the  objection  that 
just  having  the  DNA  and  bacteria  in  the  lab,  working  with  them, 
that  DNA  does  float  around,  that  there  was  a  small  chance  that  it 
would  be  taken  up  by  bacteria.   There  were  a  lot  of  ridiculous 
things . 

Hughes:   But  how  ridiculous  were  they  really  at  the  time? 

Berg:    They  were  ridiculous  only  in  the  sense  that  there  was  no  evidence 
that  there  was  a  risk.   First  of  all,  SV40  has  infected  humans. 
SVAO  was  discovered  in  the  [Salk]  polio  virus  vaccine,  so  millions 
of  kids  were  inoculated  with  SV40  and  produced  antibodies  to  SV40, 
and  there's  no  history  of  any  tumors  that  have  ever  arisen.   This 
was  documented  at  the  first  Asilomar  meeting.   That  was  one  of  the 
arguments  why  we  didn't  think  working  with  SVAO  was  so  dangerous. 

But  people  would  say,  well,  but  this  SVAO  is  coming  into  a 
human  in  a  new  way.   It's  coming  in  bacteria  that  will  essentially 
fill  the  intestinal  track  with  cells  that  are  dying  and  spilling 
out  DNA  molecules.   DNA  molecules  are  easily  destroyed,  especially 
in  an  environment  like  the  gut.   What's  the  probability  that  a  DNA 
molecule  will  survive  that?  What's  the  probability  that  DNA  will 
be  taken  up  by  an  epithelial  cell  in  the  gut?  All  these  were 
unknowns.   But  it  was  very  difficult  in  the  climate  that  I 
described  where  we  were  already  thinking  about  potential  risks. 

Second,  I  think  I  mentioned  to  you  in  one  of  our  earlier 
interviews  that  many  of  us  had  adopted  a  much  more  social 


1  D.A.  Jackson,  R.H.  Symons,  P.  Berg,  "Biochemical  method  for 
inserting  new  genetic  information  into  DNA  of  Simian  Virus  40,"  PNAS  1972, 
69:3365. 


conscience.   All  of  us  were  of  a  like  liberal  mind,  and  we  felt 
that  ethics  and  responsibility  in  science  were  important.   The 
nuclear  weapons  program  was  exploding.  All  of  us  felt  an 
obligation  not  to  do  something  that  was  involved  with  germ  warfare 
and  things  of  that  sort.   So,  there  was  a  sense  of  wanting  to  do 
the  right  thing.  And  not  doing  the  wrong  thing  because  you  were 
selfish,  because  it  was  your  experiment,  your  idea,  and  you  were 
going  to  pursue  it  hellbent;  no  matter  what  anybody  said,  you  were 
going  to  do  your  experiment.   It  was  much  more  a  matter  of  a 
social  thing:  We  ought  to  talk  this  out;  we  ought  to  think  this 
through. 

Well,  the  thinking  through  created  this  brief  interval  after 
Asilomar  I  when  nothing  happened.  And  as  Nick  Wade  and  other 
people  said,  "The  world  breathed  a  sigh  of  relief."  The  ability 
to  construct  such  DNA  molecules  was  thought  to  be  so  technically 
challenging  that  it  could  only  be  done  in  Paul  Berg's  lab  or  at 
Stanford.   And  if  they've  decided  not  to  do  this  research  anymore, 
we're  safe.   It's  interesting  how  people  misunderstood  how 
difficult  it  was  to  do.   But  what  was  true  is  people  recognized 
that  not  many  places  had  access  to  the  enzymes,  and  the  skill,  and 
the  experience  that  was  present  in  our  department.   And  so  they 
said,  "If  he's  not  going  to  do  it,  nobody  else  is  going  to  do  it." 
This  was  1971.   Less  than  two  years  later,  we  discovered  that  one 
of  these  restriction  enzymes  makes  these  cohesive  ends.   We  didn't 
have  to  synthesize  poly-A  tails  and  poly-T  tails. 


Contributions  to  Recombinant  DNA  Science 


David  Jackson's  Opinions 


Hughes:  David  Jackson  named  four  lines  of  science  which  contributed  to 
recombinant  DNA  technology.  One,  studies  of  DNA  structure  and 
physical  chemistry.1 

Berg:    We  knew  that  single  strands  that  have  complementary  sequences  form 
duplexes.   I  didn't  regard  that  as  a  major  thing,  but  it  clearly 
was  implicit  in  everything  we  did. 


1  David  A.  Jackson,  "DNA:  Template  for  an  Economic  Revolution,"  in: 
DNA:  The  Double  Helix.   Perspective  and  Prospective  at  Forty  Years,  Donald 
A.  Chambers,  ed.,  New  York:  New  York  Academy  of  Sciences,  1995,  pp.  356- 
356-365. 


95 


Hughes:   Two,  the  enzymology  of  DNA  synthesis  and  degradation;  we  talked 
about  that.   Three,  bacterial,  phage,  and  plasmid  genetics.   You 
haven't  spoken  about  plasmidology . 

Berg:    Because  we  weren't  interested  in  plasmids.   [laughter]   We  didn't 
even  know  anything  about  plasmids.   For  us,  the  plasmid  was  a 
viral  chromosome. 

Hughes:  But  plasmids  are  going  to  feed  into  [Cohen-Boyer]  recombinant  DNA. 

Berg:  Oh,  yes,  that's  the  next  stage. 

Hughes:  Four,  bacterial  restriction  and  modification  systems. 

Berg:  But  even  that  wasn't  implicit  in  anything  we  did. 

Hughes:   I  don't  think  Jackson  was  writing  specifically  about  what  the  Berg 
laboratory  was  doing. 

Berg:    In  our  case,  the  two  things  that  we  knew  were,  one,  that  in 

bacterial  systems,  viruses  could  pick  up  genes.   We  considered 
whether  that  was  also  a  possibility  in  mammalian  systems.   And  the 
second  was  cohesive  ends.   Cohesive  ends  were  a  way  to  join  DNA 
molecules  together. 

We  didn't  need  to  know  anything  about  restriction  enzymes 
because,  in  fact,  the  way  we  opened  SV40,  which  is  circular,  was 
to  cut  it  with  an  old  fashioned  enzyme,  DNase,  which  made  random 
double-strand  breaks.   That  allowed  us  to  produce  a  permuted 
population  of  linear  molecules.   They  could  be  opened  up  anywhere. 
And  that  means  what  you're  inserting  could  go  into  any  part  of  the 
SVAO  DNA  molecule.   Because,  once  we  opened  it  up,  we  could  add 
the  tails  onto  the  two  ends.   We  didn't  need  restriction  enzymes. 
So,  all  of  the  things  that  Dave  mentions  contributed  to  the 
evolution  of  the  second  stage. 

Hughes:  What  you  mean  by  the  second  stage  is-- 

Berg:  Cloning. 

Hughes:  Were  there  methods  for  joining  two  pieces  of  DNA? 

Berg:  No. 


96 
Gobind  Khorana  and  Vittorio  Sgaramella 


Hughes:   Well,  Sgaramella  was  in  Khorana 's  lab.   There  was  a  paper 

published  [1970]  on  a  ligase-mediated  form  of  DNA  joining.1  How 
does  that  work  fit  in? 

Berg:    What  Khorana  did  was  set  out  to  synthesize  a  gene  chemically.   It 
was  a  gene  that  was  about  a  hundred  base  pairs  long.  What  he 
decided  to  do  was  to  make  single-stranded  [DNA]  pieces.   Now,  if 
you  make  a  second  single-stranded  piece  that  overlaps  it,  the  two 
will  form  a  duplex  where  they  pair  and  single  strands  at  the  ends 
where  they  don't  pair.  And  then  he  made  another  single-stranded 
piece  that  paired  with  one  of  the  single  strands,  then  another 
piece,  and  so  on.   That  results  in  a  duplex  DNA  with  gaps  that  can 
be  filled  with  the  DNA  polymerase.   That  was  known;  that's  what 
Kornberg's  DNA  polymerase  I  does. 

What  Khorana  did  was  to  synthesize  a  gene  by  making  single- 
stranded  pieces  that  overlapped  each  other.  And,  whether  they 
produced  small  gaps  or  large  gaps,  they  could  all  be  filled  in. 
It  was  a  very  efficient  way  to  synthesize  a  big  piece  of  double- 
stranded  DNA,  just  to  make  overlapping  single-stranded  pieces  and 
use  the  enzymes  to  fill  in  the  rest.   You  don't  have  to  synthesize 
all  of  the  stuff  that  the  enzyme  can  fill  in. 

They  used  T4  ligase,  because  [at]  every  place  where  you  fill 
in,  you  have  ultimately  to  make  a  join.   Now,  there  were  two  DNA 
ligases,  E.  coli  DNA  ligase  and  a  similar  enzyme  that  is  encoded 
in  a  particular  bacteriophage,  T4 .   Now,  E.  coli  ligase  absolutely 
requires  that  there  be  a  complete  strand  opposite  the  site  to  be 
joined  for  joining.   But  E.  coli  ligase  cannot  join  two  pieces  of 
double-strand  DNA  at  blunt  ends.  T4  ligase  seems  to  be  able  to  do 
both  kinds  of  joins.   But  it  does  end-to-end  joining 
inefficiently.   In  other  words,  it  can  join  two  blunt-ended  pieces 
of  DNA  together.   Sgaramella  was  in  Khorana 's  lab,  and  my 
recollection  is  that  he  was  involved  in  discovering  that  T4  ligase 
could  do  blunt-end  joining. 

Hughes:   Did  they  have  the  idea  that  this  was  the  possible  mechanism  for 
joining  two  foreign  pieces  of  DNA? 

Berg:    Never  heard  anything  about  it.   Sgaramella  came  to  Stanford  and 
was  in  the  genetics  department.   He  sat  in  on  all  our  group 


1  V.  Sgaramella,  J.H.  Van  de  Sande,  H.G.  Khorana,  PNAS  1970,  67:1468- 
1475. 


97 


meetings,  so  he  knew  all  we  were  doing;  he  was  a  part  of  it.   And 
he  knew  that  T4  ligase  could  do  this,  and  we  knew  that. 

Hughes:   But  nobody  was  saying  this  was  a  possible  mechanism  for 
recombining  DNA? 

Berg:    No.   When  Sgaramella  came,  I  think  Jackson  was  already  here  and 
involved  in  the  recombinant  DNA  research.   But,  to  my  knowledge, 
Sgaramella  had  never  either  suggested  or  used  T4  [ligase]  to  join 
two  DNA  molecules  together  before  we  did  our  experiments.   In 
fact,  I  have  the  papers  here.   I've  been  carrying  them  around.   I 
wanted  at  one  point  to  write  up  something.   I  have  the  Sgaramella 
paper;  it's  1972. '  As  far  as  I  know,  Sgaramella  had  never  joined 
two  DNA  molecules  together  until  he  was  at  Stanford  and  while  he 
was  attending  our  group  research  meetings.   We  were  working  with 
SVAO  DNA,  and  he  said,  "Well,  I  can  join  P22  with  T4  [ligase]." 
Well,  the  joining  efficiency  of  TA  ligase  was  extremely  poor, 
whereas  our  joining  was  very  efficient. 

Hughes:    So  you  wouldn't  have  considered  using  his  system? 
Berg:     No,  certainly  not. 

Hughes:   It  was  never  a  discussion  point  when  Sgaramella  was  sitting  in  on 
the  meetings? 

Berg:    No;  we  were  cooking  away  on  this  experiment,  and  we  were  not 
thinking  of  using  Sgaramella 's  method. 

The  exact  chronology  of  everything  is  not  quite  as  clear  in 
my  mind  as  perhaps  it  should  be.   But  Sgaramella  was  clearly  in 
our  group  research  meetings.   He  came  frequently  and  was  there 
during  the  time  that  Jackson  was  doing  his  work.   I  think  there's 
no  question  that  [Sgaramella]  reckoned  that  instead  of  having  to 
make  cohesive  ends,  it  might  be  possible  for  TA  ligase  to  join  DNA 
molecules.   And  that  idea  grew  out  of  the  discussions  and  being 
part  of  a  common  group. 

This  is  that  little  essay  that  I  told  you  about;  as  far  as  I 
know  it  was  written  in  1969,  '70,  something  like  that.2 


1  V.  Sgaramella,  "Enzymatic  oligomerization  of  Bacteriophage  P22  DNA 
and  of  linear  Simian  Virus  40  DNA,"  PNAS  1972,  69:3389-3393. 

2  Paul  Berg,  "Can  oncogenic  viruses  be  used  to  transduce  cellular 
genes?"  [n.d.,  Berg's  personal  archive.] 


98 


Hughes:   Maybe  by  the  content  we  can  date  it. 

Berg:    No,  I  looked  through  it  again.   I  think  I  wrote  it  for  a  book, 

which  I  don't  have  a  copy  of  here,  a  compendium  on  SV40,  which  was 
being  published  at  Cold  Spring  Harbor.  We  were  asked  to 
contribute  essays  that  had  to  do  with  SV40  for  this  book,  and  so  I 
wrote  this  little  thing,  and  then  they  eventually  decided  not  to 
include  the  essays.   The  book  became  just  a  compendium  of  data 
about  SV40.   So,  1  had  this  in  my  file.   [interruption  for  lunch] 


A  Method  Difficult  to  Execute 


Berg:     Our  work  with  recombinant  DNA  raised  everybody's  consciousness 

about  making  recombinant  DNAs  and  putting  them  into  organisms  to 
do  lots  of  new  things.   But,  as  I  said,  everybody  realized  that 
the  method  we  used  was  cumbersome,  technically  challenging,  and 
maybe  not  easily  replicable. 

Hughes:   What  about  getting  the  enzymes  that  were  necessary? 

Berg:    Yes.   That's  why  I  think  everything  happened  at  Stanford.   I'm 
going  to  come  to  that  next. 


Discovery  of  Naturally  OccurrinR  Cohesive  Ends 


Janet  Mertz 


Berg:    Janet  Mertz  was  a  graduate  student  in  my  lab.   She  was  a  very 

bright,  energetic,  ambitious  kid.   I  think  she  graduated  from  MIT 
when  she  was  sixteen,  or  something  like  that,  with  a  dual  degree 
in  engineering  and  biology.   In  the  beginning  she  was  a  pain  in 
the  butt,  too. 

I  asked  her  to  identify  all  the  potentially  infectible  forms 
of  SV40.   SV40  is  a  covalently  closed,  completely  contiguous, 
circular  double-strand  DNA.   Two  strands  wind  around  each  other, 
all  the  way  around.   I  asked  her  to  determine  whether  linear  SV40 
DNA  was  infectious.   From  bacteriophage,  we  knew  that  viruses  with 
circular  DNA  genomes  were  totally  noninfectious  as  linear 
molecules.   She  was  then  to  separate  the  two  circular  strands  and 
ask  if  the  circular  strands  were  themselves  infectious  and  whether 
nicked  circles  were  infectious.   The  latter  are  made  by 


99 


introducing  a  nick  in  one  strand;  the  product  stays  together  as 
double  strands,  but  there  is  a  discontinuity  on  one  strand.   She 
started  on  this  study. 


Herbert  Boyer  and  Restriction  Enzymes 


Berg:    By  that  time,  Herb  Boyer  up  in  San  Francisco  [UCSF]  and  some  of 

his  colleagues  had  been  purifying  restriction  enzymes.   One  of  the 
enzymes  that  Herb  Boyer  purified  was  called  EcoRl.   It  came  from 
an  E.  coli  strain,  and  it  was  known  to  be  part  of  one  of  the 
restriction  modification  systems. 

Two  years  earlier,  when  I  came  back  from  La  Jolla,  I  was 
curious  about  whether  restriction  enzymes  could  be  used  to  cut 
SVAO  DNA  in  a  specific  way.  And  so,  I  got  a  couple  of  restriction 
enzymes  that  were  then  known,  isolated  by  [Matthew]  Meselson  at 
Harvard.   One  was  called  E.  coli  K,  EcoK,  and  the  other  was  EcoB. 
Francois  Cuzin,  a  postdoc  in  the  lab,  tested  them  and  found  that 
they  cleaved  SVAO  DNA,  but  at  random  sites.   Instead  of  unique 
linear  molecules  with  identical  ends,  both  enzymes  made  a 
population  of  linears  which  had  different  ends.   That  was  useless 
for  what  we  wanted,  so  I  forgot  about  it. 

When  Herb  Boyer  obtained  EcoRl,  I  asked  him  if  we  could  test 
it  on  SVAO  to  see  whether  it  would  make  unique  linears.   And  the 
answer  was,  it  did.   That  led  to  a  paper  John  Morrow  published,  in 
which  he  and  I  showed  that  EcoRl  made  a  unique  single  cut  in  SVAO 
DNA.1   It  cut  the  SVAO  once,  in  only  one  place,  and  every  linear 
was  exactly  the  same.   That  was  important  because  it  gave  us  a 
reference  point  on  the  circle.   The  point  of  publishing  the  paper 
was,  we  could  now  relate  all  other  sites  on  that  DNA  to  this  EcoRl 
site . 


1  J.F.  Morrow  and  P.  Berg,  "Cleavage  of  Simian  Virus  AO  DNA  at  a 
unique  site  by  a  bacterial  restriction  enzyme,"  PNAS  1972,  69:3365-3369. 


100 
Mertz  and  Davis:  EcoRl  Makes  Cohesive  Ends 


Berg:    In  testing  the  infectivity  of  different  forms  of  SV40  DNA,  Janet 
also  tested  the  Rl  linears  that  John  Morrow  had  made  with  Rl . 
And,  she  found  that  they  were  infectious.1 

Hughes:   It  surprised  you? 

Berg:    Big  surprise.   Not  as  infectious  as  the  circular  DNA,  but  5  or  10 
percent  as  infectious.   I  said,  "Janet,  your  linear  DNAs  are 
contaminated  with  circles.   You  didn't  get  complete  cleavage." 
So,  she  did  the  experiment  over,  and  over,  and  over  again, 
purifying  the  linear  molecules,  but  with  the  same  result. 

ft 

Berg:    So  she  began  to  look  at  them  in  the  electron  microscope,  and  sure 
enough,  they  were  all  linear.   She  didn't  find  any  contaminating 
circles  that  could  have  accounted  for  the  infectivity  she  found. 
Ron  Davis,  who  was  one  of  my  colleagues  in  the  department,  was  an 
expert  in  looking  at  DNA  molecules  by  electron  microscopy,  but 
suggested  that  maybe  they  should  look  at  the  linears  at  lower 
temperatures.   So  he  devised  a  way  in  which  you  could  mount  the 
DNA  on  the  grid  used  for  the  microscopy,  at  low  temperature.   When 
they  did  that,  many  of  the  linears  were  circles. 

Hughes:   Did  he  have  a  rationale  for  that  suggestion? 

Berg:    I  don't  know  if  he  did  or  not.   I  don't  remember  that  there  was 
any  reason  for  expecting  that  they  might  be  circles  at  low 
temperature.   I  should  say,  he  knew  that  was  the  behavior  of  these 
lambda  DNA  that  had  longer  cohesive  ends. 

As  soon  as  they  found  that,  they  realized  that  the  ends  were 
probably  cohesive,  but  very  short.  And  if  they're  very  short,  the 
stability  of  the  helix  that  you  can  form  is  going  to  be  dependent 
on  the  temperature.   So,  at  room  temperature,  things  are  linear. 
Below  4  degrees  Celsius  they  were  circular.   This  was  an 
astonishing  finding. 


1  J.E.  Mertz  and  R.W.  Davis,  "Cleavage  of  DNA  by  Rj  restriction 
endonuclease  generates  cohesive  ends,"  PNAS  1972,  69:3370-3374. 


101 


Boyer's  Group:  Sequencing  the  Cohesive  Ends 


Berg:    Well,  we  called  Herb  Boyer  and  said,  "Herb,  we've  got  this 

astonishing  thing.   These  Rl-cut  linears  have  cohesive  ends."  He 
came  flying  down  here  within  the  hour.   [laughter]   And  we  went 
through  the  data.  We  decided  that  we  were  going  to  characterize 
the  EcoRl  generated  ends,  and  he  was  going  to  sequence  the  ends. 
So  Herb  Boyer's  paper  that  Rl  made  cohesive  ends  appears  in  the 
same  issue  with  Janet  Mertz  and  Ron  Davis'  paper,  published  back- 
to-back.1  Both  these  papers  appear  in  the  same  issue  as  the 
Morrow  and  Berg  paper.   Because  PNAS  did  not  allow  an  author's 
name  to  appear  on  more  than  one  paper  in  an  issue,  my  name  does 
not  accompany  the  Mertz  and  Davis  names.   They  determine  the 
sequence  of  the  cohesive  ends  as  being  AATT. 


First  Experiment  Using  Cohesive  Ends 


Berg:     Ron  Davis  and  Janet  Mertz  performed  the  first  in  vitro 

recombination  mediated  by  the  cohesive  ends  created  by  this  enzyme 
Rl.2  It  is  a  fact  that  nobody  acknowledges  today. 

Hughes:   Why  does  nobody  acknowledge  them? 

Berg:     Because  Stanley  Cohen  prefers  not  to.   Stanley  Cohen  is  given 
credit  as  being  the  first  one  to  make  recombinant  DNAs,  and  he 
chooses  to  minimize  the  significance  of  that  [Davis-Mertz] 
finding.   Ron  and  Janet  took  two  different  DNAs,  each  with  a 
different  buoyant  density,  that  is,  when  you  centrifuge  them,  they 
separate  because  they  have  different  densities.   Both  were  cut 
with  EcoRl,  mixed  and  incubated  with  DNA  ligase  at  low 
temperature,  and  then  they  were  re-centrifuged.   Most  of  the  DNA 
now  centrifuged  at  an  intermediate  density.   That  is,  the  two 
different  DNAs  had  been  covalently  joined,  giving  molecules  with 
an  intermediate  density.   This  was  the  first  demonstration  that 
you  could  use  EcoRl  to  do  recombination  in  vitro. 

The  interesting  thing  is  that  issue  of  the  Proceedings  of 
the  National  Academy  of  Sciences  has  John  Morrow  showing  that  Rl 


1  J.  Hedgpeth,  H.M.  Goodman,  and  H.W.  Boyer,  "The  DNA  nucleotide 
sequence  restricted  by  the  Rl  endonuclease,"  PNAS  1972,  69:3448-3452. 

2  J.E.  Mertz,  R.W.  Davis,  "Cleavage  of  DNA  by  R)  restriction 
endonuclease  generates  cohesive  ends,"  PNAS  1972,  69:3370-3372. 


102 


makes  a  unique  break  in  SV40,  creating  a  reference  point.   The 
Mertz  paper  and  the  Boyer  paper  were  in  the  same  issue.   The 
Sgaramella  paper  is  also  in  that  issue  which  is  one  or  two  issues 
later  than  the  one  in  which  the  Jackson  et  al.  paper  appears.1 

Berg:     [interruption]  I  was  just  saying  that  once  it  was  clear  that  this 
enzyme  made  ends  which  were  cohesive  and  could,  in  fact,  be  used 
to  join  DNAs,  it  was  obvious  to  most  people  now  that  you  could 
join  any  DNA  molecules  together  if  they  were  each  cut  with  EcoR. 
Take  any  two  DNAs  you  want;  cut  them  with  EcoRl  to  make  sticky 
ends,  and  the  two  molecules  can  join  together. 


Peter  Lobban's  Contributions  to  Recombinant  DNA 
Thesis  Proposal,  November  1969 

Hughes:   Well,  there  are  two  other  people  that  I  think  you  should  bring 

into  this  story.   One  is  Peter  Lobban.   I  have  a  copy  of  his 

dissertation,  which  is  dated  May,  1972.   John  Lear  dates  his 
proposal  to  his  thesis  committee  to  November,  1969. 2 

Berg:     I  might  even  have  a  copy  of  it  here.   Yes,  there  it  is.3 

So  I  came  back  from  La  Jolla  in  '68  with  the  idea  to  try  to 
convert  SV40  into  a  transducing  DNA.   Dave  Jackson  was  a  graduate 
student  with  one  of  my  colleagues,  Charlie  Yanofsky,  in  the 
biology  department.   Dave  wanted  to  come  over  and  do  a  postdoc. 
He  came  over,  I  think,  in  late  1969,  but  he  had  to  finish  some 
experiments  that  he  was  doing  for  his  Ph.D.  thesis  or  a  paper  on 


1  Berg  noted  the  following:   The  Sgaramella  paper  was  submitted  to 
press  on  September  5,  1972  and  appeared  in  the  November  issue  of  that  year. 
The  work  in  the  paper  was  done  while  Sgaramella  was  at  Stanford  and 
attending  our  group  meetings.   The  Jackson,  Symons,  Berg  paper  was 
submitted  on  July  31,  1972  and  appeared  in  the  October  1972  issue  of  PNAS. 
The  Morrow-Berg  paper  was  submitted  August  16,  1972;  the  Mertz-Davis  paper 
September  11,  1972.   Both  appeared  in  the  same  issue  as  the  Sgaramella 
paper. 

2  John  Lear,  Recombinant  DNA:  The  Untold  Story,  New  York:  Crown 
Publishers,  1978,  p. 43. 

3  Peter  E.  Lobban,  "The  Generation  of  Transducing  Phage  In  Vitro," 
third  exam,  November  6,  1969. 


103 


his  thesis  work.   So,  he  didn't  get  started  on  trying  to  join  DNAs 
until  sometime  in  early  1970,  I  would  say. 

Peter  Lobban  was  a  graduate  student  with  Dale  Kaiser.   I  was 
unaware  that  he  had  presented  a  proposal  to  recombine  DMA  in  vitro 
by  using  terminal  transferase  to  create  synthetic  cohesive  ends. 
I  wasn't  on  his  proposition  committee.   Then  when  Dave  [Jackson] 
began  to  start  the  experiments,  we  found  out  that  Peter  had 
actually  proposed  the  same  approach  we  had  decided  on.   The 
concept  of  using  cohesive  ends  came  from  Dale  Kaiser,  the  same 
source  from  which  I  got  it.   Dale  Kaiser  was  actively  working  on 
lambda  cohesive  ends,  and  Peter  Lobban,  in  his  research  group,  was 
clearly  motivated  by  that,  as  was  I.   Normally,  these  Ph.D. 
propositions  are  exercises  in  which  people  present  an  idea. 
They're  supposed  to  demonstrate  a  capacity  for  being  creative  in 
generating  a  new  idea,  devising  an  experimental  way  of  testing 
their  idea,  and  then  they  continue  on  their  thesis,  whatever  that 
is.   Most  often,  the  proposal  and  thesis  are  totally  unrelated. 


Lobban 's  Communication  with  the  Berg  Group 


Berg:    1  guess  Peter's  committee,  or  Peter  himself  and  Dale  Kaiser,  were 
so  struck  by  the  novelty  of  the  idea  that  Peter  dropped  what  he 
was  doing  on  his  thesis,  and  he  started  to  try  to  do  what  he 
described.   My  recollection  is  that  in  the  beginning,  unbeknownst 
to  me,  when  Dave  Jackson  started  on  it,  as  is  often  the  case,  the 
people  who  were  at  the  bench  talked  more  to  each  other  than  I 
talked  to  Peter  Lobban.   And  Dave  found  out  that  Peter  Lobban  was 
also  trying  to  develop  the  same  technique.   The  two  of  them,  if 
they  didn't  actually  collaborate,  were  in  very  close 
communication.   What  was  very  interesting  was  how  easy  the 
communication  was. 


Lobban 's  Discoveries  and  Speculations  for  Practical 
Application 

Berg:    Peter,  and  we  actually  refer  to  him  in  our  paper,  made  several 

critical  discoveries  that  facilitated  the  technique.   For  example, 
when  you  cut  DNA  and  both  ends  are  either  flush  or  very  close  to 
being  flush,  it's  very  difficult  for  terminal  transferase  to  add 
nucleotides  onto  the  end.   On  the  other  hand,  if  you  peel  back  the 
5-prime  strand,  and  you  have  the  3-prime  end  sticking  out,  then 
this  enzyme  adds  nucleotides  very  well.   Peter  discovered  that. 


104 


He  very  quickly  communicated  that  to  us,  so  that  if  we  were  going 
to  make  cohesive  ends,  it  was  better  to  pre-treat  the  DNA  with  an 
enzyme  that  cuts  back  the  5-prime  end  a  little  bit;  we 
acknowledged  Peter's  contribution  in  our  paper. 

And  so,  there  was  a  very  close  communication,  but  it  wasn't 
a  collaboration.  We  never  intended  to  be  working  on  the  same 
project.   Since  I'd  never  seen  his  proposition,  I  was  unaware  of 
the  implications  of  what  he  foresaw  for  being  able  to  do  this. 
Our  research  was  strictly  motivated  by  trying  to  get  DNAs  to 
attach  to  SV40  so  we  could  introduce  them  to  mammalian  cells. 

Hughes:   What  does  Lobban  speculate? 

Berg:     I  think  he  begins  to  see  this  as  gene  therapy.   It  has  been  a  long 
time  since  I've  read  his  proposal.   [Reading  from  a  copy  supplied 
by  Hughes]:  "An  eventual  goal  for  the  method,  assuming  its 
success,  a  simple  task  would  be  to  produce  a  collection  of 
transductants  synthesizing  the  products  of  genes  of  higher 
organisms."   In  other  words,  he  would  attach  something  that  would 
get  it  into  E.  coli.   "This  would  be  of  immense  help  in  the 
purification  of  proteins  made  by  these  genes,  for  in  many  cases 
they  would  be  far  less  dilute  in  the  bacterium  than  in  the  cells 
of  their  origin."   So  he  begins  to  see  that  it's  possible  to 
manufacture  the  products  of  genes,  which  ultimately  came  on  much 
later. 

"It  is  even  conceivable  that  transduction  can  be  used  for 
fine  structure  mapping  of  the  genes  they  bear.   For  phage  can  be 
mutagenized,  and  a  collection  of  point  mutants  could  be  obtained." 
He  was  already  visualizing  that  you  could  begin  to  make  mutations 
in  whatever  you  made.  And,  if  you  put  it  back  into  cells,  you 
could  determine  whether  the  mutations  affected  the  function,  and 
so  on. 

Hughes:   In  the  dissertation,  he  mentions  the  possible  uses  of  this 

technology,  and,  of  course,  the  main  one  is  for  transduction.   He 
talks  about  the  DNAs  being  used  "in  a  search  for  proteins  and 
other  macromolecules  present  in  mammalian  cells  that  might 
interact  with  DNA  in  a  gene- specif ic  manner  and  thus  be  involved 
in  control  of  gene  expression."1  And  he  talks  about  fine 
structure  mapping.   "[The  method]  could  be  used  as  a  source  of  the 


1  Peter  Edward  Lobban,  "An  enzymatic  method  for  end-to-end  joining  of 
DNA  molecules,"  Dissertation,  May  1972.  (Lane  Medical  Library,  Stanford, 
dissertation  #3781  1972L) 


105 


gene  products  that  might  be  far  more  convenient  than  the  mammalian 
cells  themselves."1 

Berg:    Yes,  that's  what's  in  this  proposal. 


Not  Cloning 


Hughes:   So,  Lobban  has  the  idea  of  cloning. 

Berg:    Well,  he  doesn't  have  the  idea  of  cloning.   I  think  what  he  has 
the  idea  of  is  constructing  molecules  which  could  then  be 
introduced  into  cells  that  would  express  the  transduced  segment, 


The  Cohen-Boyer  DNA  Cloning  Experiments 

Berg:    But  the  cloning  really  was  a  very  different  procedure.   As  far  as 
I  remember,  the  first  real  notion  of  cloning  comes  out  of  the 
experiment  that  John  Morrow  did  together  with  Herb  Boyer  and 
Stanley  Cohen  where  they  cloned  the  ribosomal  DNA  sequences  from 
the  frog.2 

Hughes:   The  very  first  cloning  experiment  was  published  in  1973,  using-- 
Berg:     pSClOl.3   Stan  joined  it  to  another  plasmid. 


1  Ibid. 

2  J.F.  Morrow,  S.N.  Cohen,  A.C.Y.  Chang,  H.W.  Boyer,  H.M.  Goodman, 
R.B.  Helling,  "Replication  and  transcription  of  eukaryotic  DNA  in 
Escherichia  coli,"  Proceedings  of  the  National  Academy  of  Sciences  197A, 
71:1743-1747. 

3  S.N.  Cohen,  A.C.Y.  Chang,  H.W.  Boyer,  R.B.  Helling,  "Construction  of 
biologically  functional  bacterial  plasmids  in  vitro,"  Proceedings  of  the 
National  Academy  of  Sciences  1973,  70:3240-3244. 


106 


Hughes:  There  is  a  paper,  which  precedes  the  Xenopus  [frog]  paper,  in 
which  Cohen  speculates  on  the  practical  possibilities  of  this 
procedure. ' 

Berg:    My  recollection  is  that  the  introduction  of  a  foreign,  totally 

unrelated  DNA,  frog  DNA,  into  a  bacterial  plasmid,  introducing  it 
into  bacteria  and  getting  the  bacteria  to  propagate  these  was  the 
first  demonstration  that  you  could  propagate  foreign  DNA  in 
bacteria.   And  two,  that  each  colony  produced  from  this  was  a 
clone,  a  clone  of  the  original  DNA. 

In  fact,  the  Morrow  experiment  starts  with  a  piece  of  DNA 
which  is  a  mixture,  not  a  unique  segment  of  DNA,  and  therefore 
each  colony  represents  only  one  component  of  that  mixture.   That's 
cloning.   Now,  what  I  remember  that  Stan  Cohen  did  was  to  take  a 
Staphylococcus  aureus  plasmid,  fuse  it  to  the  E.  coli  plasmid,  and 
show  that  it  can  introduce  the  drug  resistance  property  of  the 
Staph  aureus  plasmid  to  E.  coli. 

Hughes:   But,  there's  no  talk  about  cloning? 

Berg:  I'd  have  to  go  back  and  read  the  paper  in  detail  to  know.  I 
believe  that  the  big  breakthrough  was  the  Morrow  experiment, 
showing  that  you  can  actually  clone  foreign  DNAs.  That's  my 
recollection. 


More  on  Recombinant  DNA 


More  on  Lobban-Berg  Group  Interactions 


Berg:     Now,  to  go  back  to  Peter  Lobban.   Peter  and  we  decided  that  we 

should  try  to  publish  together.   Then,  at  one  point,  Peter  said, 
"No,  I  don't  want  to  publish  in  the  PNAS  [Proceedings  of  the 
National  Academy  of  Sciences],  I  want  to  publish  it  in  JMB 
[Journal  of  Molecular  Biology],  And,  I  would  rather  wait." 


1  A.C.Y.  Chang  and  S.N.  Cohen,  "Genome  construction  between  bacterial 
species  in  vitro:  Replication  and  expression  of  Staphylococcus  plasmid 
genes  in  Escherichia  coli,"  Proceedings  of  the  National  Academy  of  Sciences 
1974,  71:1030-1034. 


107 


Hughes:   I've  read  a  slightly  different  account.1   Dale  Kaiser  wasn't  quite 
satisfied  and  wanted  Peter  to  do  more  research. 

Berg:    Could  be;  I  don't  remember.   They  knew  we  were  going  to  publish. 
There  was  never  any  question  that  we  sneaked  in  a  publication; 
they  knew  we  were  going  to  publish  it  in  PNAS  and  when. 

Hughes:   And  you  each  cite  each  other.   1  noticed  that  as  well. 

Berg:    Oh,  yes.   It  was  kind  of  a  friendly  and  collaborative  program, 
with  the  understanding  that  we  were  each  trying  to  do  something 
different,  but  using  the  same  idea.   I  believe  we  hit  on  this  idea 
independently--!  have  no  doubt  about  that—because  I  was  totally 
unaware  of  Peter's  having  presented  his  proposal. 

We  had  a  visit  from  Jim  Wang  from  Harvard.   He  reported  that 
he  found  two  circular  DNA  molecules  called  concatamers  intertwined 
in  certain  cells.   And,  when  I  heard  his  seminar,  which  would  have 
been  probably  late  '68,  early  '69,  I  thought  maybe  that  was  the 
way  we  could  get  DNA  into  mammalian  cells.   That  is,  we  would  use 
SVAO,  which  is  circular,  and  we  would  try  to  take  foreign  DNA  and 
circularize  it  with  SVAO  to  form  concatamers.   If  the  SVAO  DNA 
went  into  cells,  it  would  carry  with  it  this  foreign  DNA.   Well, 
we  tried  different  ways  to  generate  such  concatamers,  and  we 
failed.   So,  then  we  decided,  well,  maybe  another  way  to  do  it  is 
to  open  up  the  SVAO  ring  and  join  DNA  segments  to  it  by  making 
cohesive  ends.   And  so  that's  sort  of  the  genesis  of  our  idea. 

It  wasn't  so  novel  because  we  knew  that  cohesive  ends 
worked.   And  we  knew  that  there  was  a  way  to  make  cohesive  ends. 
And  so  that's  what  I  laid  out  for  Dave  Jackson  as  a  postdoc 
project.   But  he  had  to  finish  up  research  in  progress  in 
Yanof sky's  lab,  and  he  was  delayed  in  getting  started.  Once  he  got 
started,  he  discovered  that  Peter  Lobban  had  come  up  with  the  same 
idea  and  was  working  on  it.   And  then,  of  course,  we  realized  that 
we  were  both  trying  to  do  the  same  things,  but  for  different 
purposes.   We  had  a  specific  purpose  in  mind.   Peter  was  more 
interested  in  a  general  method  for  joining  DNA  molecules.   We  were 
less  interested  in  the  way  to  join  DNA  molecules  as  making 
molecules  that  we  could  use. 


1  Peter  Lobban  to  Arthur  Kornberg,  October  10,  1986  (Kornberg's 
personal  correspondence). 


108 


Berg  Questions  Lobban's  Use  of  Two  Identical  Molecules 


Hughes:   Isn't  your  different  goal  indicated  by  what  you  chose  as  your 
system.   Namely,  you  were  using  SVAO  and  lambda  dv  gal. 

Berg:    I  don't  remember  what  Peter  used. 
Hughes:   He  used  two  p22  molecules. 

Berg:     Yes,  that's  right.   My  own  view  was  that,  if  you  want  to  develop  a 
method  to  do  something,  you'd  want  to  do  it  with  things  that  you 
could  then  do  the  next  step.   What  was  the  endpoint  of  joining  two 
p22  molecules  together? 

Hughes:   Well,  I  thought  about  that,  too.   I  thought,  well,  here's  a 

graduate  student  trying  to  pick  a  circumscribed  problem.   He's 
just  worried,  can  he  join  two  molecules?   What  the  molecules  are 
doesn't  matter  to  him. 

Berg:     Yes,  but  he  writes  and  projects  multiple  uses  and  potential. 

Hughes:   Well,  I  know  he  does.   I  guess  you  could  still  say,  why  not  use 
two  different  molecules? 

Berg:    Why  not  try  it  out  with  the  molecules  you  ultimately  would  like  to 
see  some  function  for? 

Hughes:   Maybe  there's  something  in  the  dissertation  about  something 

physically  favorable  about  using  two  p22  molecules.   I  didn't  copy 
the  whole  dissertation. 

Berg:     [scans  dissertation]   Yes,  see:   He  refers  to  Sgaramella  here-- 
"Sgaramella  personal  communication,  May  '72."   "So  far  the 
enzyme"--T4  ligase--"has  been  used  to  join  certain  small  synthetic 
DNA  molecules  and  to  make  linear  dimers  and  trimers  of  P22 
DNA...but  no  circles.   The  advantage  of  the  ligase-mediated 
joining  reaction  is  it's  simplicity,  for  whenever  the  molecules  to 
be  joined  have  base-paired  ends,  only  a  single  reaction  is 
required.   However,  there  is  no  way  to  ensure  that  a  given  DNA 
molecule  will  not  join  with  another  molecule  of  the  same  type..." 
Whereas,  when  you  make  cohesive  ends,  you  direct  who's  going  to 
join  with  who. 

Hughes:   Yes,  exactly.   But  we  haven't  answered  why  he  chose  p22. 

Berg:    I  don't  know.   I  thought  at  the  time- -I  don't  know  that  I  thought 
very  deeply  about  it- -but  I  couldn't  understand  why  he  was 
spending  time  with  p22,  anymore  than  why  Sgaramella  used  p22.   We 


109 

wanted  to  develop  a  joining  method  to  actually  construct  something 
that  we  could  test.   That  may  not  sound  very  profound-- 

Hughes:   I  understand  what  you're  getting  at. 

Berg:    In  fact,  today,  if  someone  develops  a  technique  or  a  method  and 
tries  to  publish  it,  we  ask,  "What  question  are  you  trying  to 
answer?"   It  would  have  to  be  some  dramatic  new  technology  that's 
going  to  open  up  a  whole  new  world  before  a  journal  will  actually 
publish  something  on  a  technique,  and  even  then  it's  hard  [to  get 
it  published].   But,  if  you  do  it  in  the  context  of  answering  a 
question,  you  develop  a  way  to  test  or  answer  a  new  question,  then 
the  journals  are  more  willing  to  accept  the  paper. 


The  Jackson,  Symons ,  Berg  Paper,  October  1972 


Berg:     That's  what  we  thought  we  were  doing,  and  that's  what  we  outlined 
in  our  paper.   We  certainly  visualized  some  of  the  things  that 
could  be  done  with  that  approach.   I  can't  remember  whether  our 
paper  actually  suggests  growing  SV40  in  E.  coll.   That  paper 
exceeded  the  length  limits  of  the  PNAS  by  one  page.   They  in  fact 
accepted  it,  for  very  special  reasons,  they  said.   But,  it  allowed 
us  to  discuss  the  implications. 

Hughes:   What  were  the  special  reasons? 

Berg:     They  recognized  that  this  was  a  breakthrough.   In  fact,  I  think 

there  was  an  editorial  in  Nature  about  it.   So,  it  was  clear  that 
right  away  the  world  saw  that  the  ability  to  join  DNA  molecules 
together  in  vitro  opened  up  a  whole  new  direction. 


Lobban's  Work  Ignored 


Berg:    And  I  think  much  to  the  dismay--!  shouldn't  say  dismay,  but 

certainly  it  was  unfortunate—Peter  Lobban's  work  got  very  little 
attention.   To  this  day,  a  lot  of  people  look  back  and  don't 
understand  why,  except  if  you  know  Peter  Lobban.   He's  very,  very 
low-key,  very  phlegmatic.   He  went  off  to  Canada,  did  a  postdoc  in 
a  very  different  area,  cell  biology.   Then  he  began  to  interview 
for  jobs,  and  [spoken  with  pause  between  each  word:]  did  -  not  - 
get  -  any  -  offers  -  for  -  jobs.   He's  a  remarkably  bright  guy. 
He  gave  up  and  got  a  job  and  went  back  to  engineering  school.   And 
I  think,  for  people  who  knew  Peter,  that's  his  bent.   His  bent  is 


110 

inventing  new  things  and  applying  them.   He  now  works  for  a 
company  of  which  I'm  a  director. 

Hughes:   Small  world. 

Berg:    He  works  on  the  engineering  aspect  of  this  company's  project. 

Hughes:   I'll  look  up  the  date  you  submitted  the  Jackson,  Symons,  Berg 
paper.  [July  31,  1972] 

You  wrote  a  letter  to  Hilary  Koprowski  in  May  of  1973  in 
which  you  mentioned  your  relationship  with  Peter:1  "...at  all 
times  we  had  a  very  amicable,  cooperative  and  most  important 
complementing  "collaboration",  so  much  so,  that  it  was  Peter  who 
often  made  the  key  breakthroughs  to  solving  technical  problems." 
That's  an  almost  contemporary  account  of  what  you  had  already  told 
me . 

Berg:     Yes,  almost  the  same  words. 


Recombinant  DNA:  Jensen,  Wodzinski,  and  Rogoff,  1971  If 


Hughes:   In  his  dissertation,  Lobban  mentions  a  paper  by  Jensen  and 
colleagues ,2 

Berg:    I'd  never  heard  of  him.   Stan  Cohen  wrote  me  a  letter  after  that 
Philadelphia  meeting  in  which  he  tells  me  about  Jensen.   He  might 
have  mentioned  it  in  his  talk  at  the  Beckman  Center  History 
Library  Symposium.   But  I  frankly  had  never  heard  of  Jensen 
before. 

Hughes:   Well,  Lobban  knew  about  him.   Here  it  is.   [Indicates  page  in 

Lobban  dissertation].   Read  it  aloud,  if  you  don't  mind,  for  the 
tape. 

Berg:     Continuing  from  Lobban 's  thesis,  "During  the  course  of  these 
experiments,  a  paper  appeared  in  the  literature  (Jensen, 
Wodzinski,  and  Rogoff,  1971)  describing  an  attempt  to  join 


1  Berg  to  Koprowski,  May  22,1973  (Berg  papers,  SC358,  Green  Library, 
Stanford,  box  3,  folder : 1973) . 

2R.H.  Jensen,  R.J.  Wodzinski,  M.H.  Rogoff,  "Enzymatic  addition  of 
cohesive  ends  to  T7  DNA,"  Biochemical  and  Biophysical  Research 
Communications  1971,  A3,  no. 2:384-392. 


Ill 

molecules  of  the  DNA  of  T7  phage  together  by  a  method  that  called 
for  the  use  of  terminal  transf erase  to  create  cohesive  ends.   The 
conclusions  reached  by  the  authors  were  much  more  pessimistic  than 
those  stated  here.   They  made  what,  in  our  symbols,  would  be  dA- 
T7-dA  and  dT-T7-dT,  but  when  the  two  DNA's  were  annealed,  joining 
was  relatively  poor,  and  no  circles  were  seen  even  though  linear 
dimers  and  trimers  were  formed.   It  would  appear,  then,  that  in 
their  hands  terminal  transferase  did  not  add  homopolymer  blocks  to 
both  ends  of  very  many  of  the  molecules;  the  reason  is  that  they 
did  not  treat  the  T7  DNA  with  lambda  exonuclease  prior  to  using 
the  transferase."  That's  in  order  to  peel  back  the  end. 

Hughes:   I  see. 

Berg:     [continuing  to  read  from  Lobban's  thesis]  "In  our  hands,  T7  DNA 

primes  the  transferase  reaction  with  the  same  kinetics  as  P22  DNA; 
that  is,  there  is  an  acceleration  phase.   The  authors  also  were 
unable  to  close  their  joined  molecules  with  ligase  alone  or  with 
ligase  with  polymerase;  the  reasons  for  that  problem  are  not 
obvious . '" 

I  don't  remember  ever  seeing  that  paper  by  Jensen  or  hearing 
anybody  even  discuss  it.   I  can't  believe  it's  blocked  out  [of  my 
memory]  completely,  but  I  don't  remember  ever  hearing  about  it. 
In  1971,  we  were  well  along  on  the  work.   If  Peter  knew  about  [the 
Jensen  et  al.  paper],  it  would  be  surprising  that  Dave  Jackson 
would  not  have  known  about  it.   But  certainly,  I  don't  remember 
ever  hearing  anything  about  it. 


Recombinant  DNA  Construction  Using  Terminal  Transferase 


Berg:    Well,  actually,  there  was  another  person  who  was  trying  to  do  the 
same  thing  whom  I  knew  but  didn't  know  about  his  work.   I  went  to 
give  a  seminar  at  Merck  where  I  described  this  recombinant  work. 
This  guy,  who  was  working  at  Merck  [and]  had  been  a  student  of  one 
of  my  friends,  told  me  that  he  had  come  up  with  this  same  idea  of 
constructing  recombinants  using  terminal  transferase.   The  people 
in  power  at  Merck  who  decide  on  what  people  work  on  said,  okay,  he 
could  give  it  a  try.   He  worked  on  it;  he  couldn't  get  it  to  work 
for  about  four,  five,  six  months  or  something,  and  they  ordered 
him  to  give  up  the  project. 


Lobban  dissertation,  May  1972,  pp.  120-121 


112 


It  was  only  several  months  later  that  I  came  to  give  the 
seminar  and  described  the  successful  use  of  that  approach.   He  was 
not  devastated  but  he  was  certainly,  as  you  might  expect,  kicking 
somebody  for  having  prevented  him  from  working  on  it  longer.   The 
surprising  thing  is  it  was  actually  easy,  but  he  did  not  have 
access  to  the  same  enzymes,  although  he  had  worked  in  a  DNA 
replication  lab.   He  had  gotten  his  degree  with  Jerry  Hurwitz,  so 
he  knew  how  to  use  enzymes.   I'm  blanking  on  his  name.   Fred 
something. ' 


David  Hogness:  Cloning  of  Eukaryotic  DNA 


Hughes:   I  read  that  David  Hogness  very  early  began  to  use  the  technique 
with  eukaryotic  DNA. 

Berg:     Dave  used  the  technique  of  dAT  joining.   My  recollection  is  that 
he  did  it  with  plasmids.   He  wanted  to  clone  Drosophila  DNA.   He 
wanted  to  isolate  segments  of  the  Drosophila  genome.   If  you  cut 
DNA  with  EcoRl,  you  can  only  cut  it  in  defined  locations,  at 
sequences  that  the  enzyme  recognizes.   But,  if  you  want  to  make 
random  breaks  so  you  get  all  possible  sequences,  then  you  make 
other  kinds  of  breaks.   You  shear  the  DNA.   If  you  run  it  through 
a  Waring  blender,  the  forces  cause  the  DNA  to  break,  and  they 
break  at  random  sites.   Now,  if  you  do  that,  you  don't  have 
cohesive  ends.   So  then  you  have  to  build  the  cohesive  ends. 

But,  this  was  after  Morrow,  Cohen,  and  Boyer  showed  that  you 
could  actually  clone  a  foreign  piece  of  DNA.   So  Hogness  said, 
"Okay,  I'm  going  to  use  a  plasmid.   I'm  going  to  make  AT  ends  on 
the  plasmids,  and  I'm  going  to  take  Drosophila  DNA,  shear  it  down 
to  a  distribution  of  sizes.   I'm  going  to  put  A's  or  T's  on  it, 
and  then  I'm  going  to  clone  it,  which  is  what  he  did. 

And  so,  he  was  able  to  clone,  for  the  first  time,  bits  of 
Drosophila  DNA  in  bacteria.   I  don't  think  many  people  outside  of 
Stanford  used  that  approach.  Well,  I'm  not  sure.   There  could 
have  been  some  other  people  who  used  AT  joining.   But  not  too  many 
people  used  AT  joining  because  they  just  thought  that  it  was 
cumbersome,  difficult. 


In  editing,  Berg  could  not  recall  the  scientist's  last  name. 


113 


Discovery  of  Other  Restriction  Enzymes  Making  Cohesive  Ends 

Berg:    And  then,  it  was  discovered  that  other  restriction  enzymes  also 

made  cohesive  ends.   We  began  to  develop  a  battery  of  ways  to  cut 
DNA  leaving  cohesive  ends.   If  you  use  BemHl,  another  enzyme,  it 
cuts  DNA  but  creates  different  cohesive  ends.   If  you  cut  the 
plasmid  with  the  same  enzyme,  then  you  can  join  those  together. 
As  the  number  of  restriction  enzymes  began  to  increase  very 
quickly,  we  learned  which  ones  produced  cohesive  ends,  which  ones 
didn't.   So  now  you  have  a  whole  battery  of  tools  to  be  able  to 
join  different  DNAs  together.   You  want  to  restrict  it  by  saying, 
"I'm  only  going  to  cut  the  DNA  at  Rl  sites." 


Stanley  N.  Cohen  and  the  Cloning  Experiments 

Departmental  Affiliation  and  Early  Research  Interest 


Hughes:   You  had  a  relationship  with  Herb  Boyer  which  predated  his 
recombinant  DNA  experiments.   And  was  Stan  Cohen--? 

Berg:     Stan  Cohen  was  in  the  Department  of  Medicine. 
Hughes:   Not  in  Genetics  at  that  point?1 

Berg:     He  was  in  the  Department  of  Medicine.   I  think  he  had  a  joint 

appointment.   But,  he  was  primarily  the  head  of  the  Division  of 
Clinical  Pharmacology  in  the  Department  of  Medicine.   Because  I 
was  very  close  friends  with  Jerry  Hurwitz  with  whom  Stan  was  a 
postdoc,  I  knew  all  about  Stan  Cohen.   When  he  came  to  Stanford, 
he  decided  to  work  on  plasmids  because  plasmids  were  important  in 
clinical  medicine.   It  was  clear  that  plasmids  were  the  carriers 
of  drug-resistance  genes,  and  drug-resistance  was  a  big  problem  in 
clinical  medicine,  and  so  he  was  studying  the  carriers  of  drug- 
resistance.   Stan  did  a  very  nice  job  of  characterizing  the 
circular  DNA  molecules.   If  you  ask  where  does  the  person  who  has 
those  kinds  of  interests  find  their  intellectual  mates,  it  is  in 
biochemistry.   And  so,  he  hung  around  in  [the  Department  of] 
Biochemistry  most  of  the  time. 


Cohen  joined  the  Department  of  Genetics  in  1978. 


114 


Mort  Handel's  Procedure  for  Introducing  DNA  into  Bacteria 


Berg:    When  he  was  isolating  plasmids,  one  of  the  experiments  he  wanted 
to  do  was  to  get  plasmids  back  into  bacteria.   And  there  was  a 
postdoc,  or  sabbatical  visitor,  in  Dale  Kaiser's  lab.   Mort-- 

Hughes:   Mandel? 

Berg:    Mandel  had  discovered  that  if  you  took  E.  coli  and  exposed  them  to 
elevated  calcium  and  gave  them  a  shock,  DNA  entered  these  cells 
much,  much  more  frequently  and  efficiently  than  if  you  didn't  do 
that.   And  so,  when  Stan  heard  that,  he  came  around  and  wanted  to 
learn  how  to  do  this.   Now,  Janet  Mertz,  who  was  in  my  lab,  was 
already  doing  that.  And  so  she  instructed  Stan  on  how  to  do  this, 
and  they  actually  worked  together. 


Berg's  View  of  the  Genesis  of  the  Cohen-Boyer  Experiments 

Berg:     When  the  Rl  experiment  showed  that  the  existence  of  cohesive  ends 
could  be  used  to  join  DNAs,  Stan  immediately  grasped  the  relevance 
of  that  as  something  he  could  do.   And  he  said,  "Okay,  if  that's 
true,  I  can  take  two  different  plasmids,  cut  them  with  Rl,  anneal 
them  together,  ligate  them,  and  then  see  if  I  can  put  them  back 
into  cells.   Janet  was  helping  him  with  the  technology  of 
reintroducing  DNA  into  cells. 

We  were  aware  of  those  experiments.   Herb  Boyer  was  not 
involved  at  all  as  far  as  I  know.   Then  the  story  goes  that  Cohen 
and  Boyer  were  at  a  meeting  in  Hawaii,  and  they  were  sharing 
pastrami  sandwiches,  and  they  had  this  great  idea  that  maybe  they 
could  in  fact  reconstruct  new  kinds  of  plasmids  and  put  them  back 
into  bacterial  cells.   If  I  remember,  Stan  had  just  taken  pSClOl, 
cut  it,  and  shown  that  you  could  make  dimers.   But  now  the 
discussion  with  Boyer  was  to  do  this  experiment  of  joining  two 
different  DNAs  together,  and  they  did  that.   I'm  not  sure  who  in 
Stan  Cohen's  lab  did  that  experiment. 


John  Morrow  and  the  Xenopus  Experiment 


Berg:    At  the  same  time,  John  Morrow  was  finishing  up  his  Ph.D.  in  my 

lab.   And  he  was  going  to  do  a  postdoc  with  Don  Brown  at  Carnegie. 
Brown  had  sent  him  some  frog  ribosomal  DNA,  for  what  reason  I 


115 

don't  know.   But  it  was  in  Morrow's  refrigerator.   Unbeknownst  to 
me,  and  without  him  telling  me,  Morrow  went  to  Stan  Cohen  and 
Boyer  and  said,  "We  ought  to  be  able  to  introduce  this  frog  DNA 
into  E.  coli  by  linking  it  to  the  plasmid." 

Hughes:   You  told  me  that  this  was  ribosomal  DNA.   They  were  using  this 
particular  DNA  because-- 

Berg:    It  was  foreign.   John  happened  to  have  it  in  his  refrigerator.   I 
suppose  you  could  have  asked,  if  I  put  it  into  bacteria,  would 
this  frog  ribosomal  DNA  be  expressed?  And  maybe  that  was  on  his 
mind.   But,  John  never  told  me  anything  about  this.   He  kept 
telling  me  that  he  was  delayed  in  finishing  his  thesis  because  of 
computer  problems,  or  this  problem,  or  that  thing,  all  of  which 
was  beginning  to  be  very  fishy. 

Hughes:   Were  you  suspicious  at  the  time? 

Berg:     1  wasn't  suspicious  at  the  time.   I  had  no  idea.   People  in  the 
lab  knew,  but  nobody  said  anything  to  me  about  it.   Eventually, 
the  experiment  was  done,  and  John  came  to  me  and  told  me  about  it. 
I  almost  kicked  him  out  of  the  lab,  I  was  so  furious.   He  was 
using  me  and  lying  to  me  about  what  was  delaying  his  departure. 
In  fact,  surreptitiously,  he  had  gone  off--   He  had  every  right  to 
do  that,  but  at  least  he  could  have  been  upfront  about  it. 

Well,  years  later,  I've  gotten  many  letters  from  John  Morrow 
reminiscing  about  that  period  and  how  remorseful  he  had  been  and 
how  much  he  credits  me  with  not  having  destroyed  his  career.   The 
fact  is,  our  department  went  off  on  its  retreat.   He  remained 
behind  because  he  was  literally  finished,  and  people  who  are 
finishing  up  their  thesis  usually  don't  come  to  our  retreats.   But 
because  I  thought  the  experiment  was  so  terrific,  I  called  him  and 
invited  him  to  come  and  give  a  talk  to  the  department  on  this 
experiment.   He  drove  down  to  Asilomar  and  gave  the  talk.   And  he 
has  acknowledged  that.   The  point  is,  I  did  not  know  about  that 
experiment  when  it  was  being  conducted.   I  regard  it  as  one  of  the 
critical  experiments  in  the  whole  evolution  of  the  DNA  cloning 
technology.   Stan  and  Herb  either  ignore  it  or  fail  to  give  it 
sufficient  credit. 


Morrow  and  Helling  Challenge  Patent  Inventorship 


Berg:    Regarding  the  Cohen-Boyer  cloning  patent,  John  Morrow  hired 

lawyers  and  sued  them  because  they  excluded  him  from  the  patent, 


116 


which  essentially  was  based  on  cloning.   And  Helling  also  sued 
them. 

Hughes:   But  they  remain  off  the  patent. 

Berg:     Yes,  that's  right.   They  were  told  that  patent  law  says  it  can't 
have  more  than  two  people  as  inventors;  I  don't  even  know  if 
that's  true  [it's  not].1  But,  in  any  case,  John  was  resentful  for 
a  long,  long  period  afterwards.   He  was  screwed  out  of  any  of  the 
benefits  from  that  or  the  recognition  of  it. 


Berg  Claims  Two  Key  Experiments  in  Cloning  DNA 

Berg:     So,  I  regard  that  two  key  experiments  were  done  in  our  department; 
they  were  both  done  by  my  students,  but  not  necessarily  under  my 
supervision:  Janet  clearly  discovered  the  cohesive  ends  of  Rl--she 
and  Ron  Davis--and  showed  for  the  first  time  that  they  could  be 
used  to  join  DNA  molecules  together.   And  John  Morrow  was  the 
first  one  to  clone  defined  DNA  sequence  in  E.  coli  and  by  that 
experiment  establish  the  concept  of  cloning.   And  neither  one  of 
them  is  widely  acknowledged.   People  who  know  recognize  what  they 
did.   But  they  never  have  gotten  the  kind  of  credit  that  Boyer  and 
Cohen  got.   Well,  once  the  experiment  was  done  with  Xenopus  DNA, 
it  became  pretty  clear  that  you  could  put  any  DNA  into  E.  coli. 
And,  that's  when  the  whole  thing  exploded. 


The  Commercial  Potential  of  Cloning  Technology 
Berg  Doesn't  Hear  of  It 


Hughes:   Were  you  thinking  about  the  commercial  potential  of  this 
technology,  before  the  Xenopus  experiment? 

Berg:    Not  me;  1  never  heard  anybody  talk  about  it.   That's  why  I  would 

say  it's  sort  of  a  surprise  to  see  it  in  Peter's  proposal,  because 
I  had  never  seen  this  proposal.   I'd  never  heard  anybody  talk 
about  potential  commercial  value. 


1  For  the  history  of  this  major  patent,  see:   Sally  Smith  Hughes,  "The 
Cohen-Boyer  Recombinant  DNA  Cloning  Patent  and  the  Accelerating 
Commercialization  of  Academic  Biology"  (in  press). 


117 


Genentech  and  Cetus  Palo  Alto 

Berg:    The  first  I  heard  of  commercial  discussions  was  that  Genentech  was 
being  formed,  largely  through  Herb  Boyer.   I  think  in  those  days 
most  of  us  who  were  academics  were  somewhat  disdainful  of 
commercial  involvement.   In  fact,  it  created  a  lot  of  problems 
with  UCSF,  enormous  angst:  Who  did  what?  And  who  owned  what?  And 
so  on  and  so  forth.   There  were  probably  suits  between  UCSF  and 
Genentech  about  materials.   Genentech  didn't  have  a  place  to  work. 
So  Herb  Boyer,  who  was  "Genentech",  was  working  at  UCSF 
laboratories,  and  that,  most  of  us  thought,  was  totally  improper. 
And  then,  not  long  after,  Cetus  actually  formed  a  laboratory  for 
Stan  Cohen.   It  was  called  Cetus  Palo  Alto,  but  after  some  time  it 
failed. 

Hughes:   Why  did  it  fail? 

Berg:    Can  we  turn  that  recorder  off?   [interruption] 


Berg's  Nobel  Prize  Address;  Citation  of  Cohen-Boyer  Research 


Hughes:   I  read  your  Nobel  address,  in  which  you  cite  a  lot  of  work,  but 
you  don't  cite  the  Cohen  and  Boyer  papers.1 

Berg:    My  recollection  is  that  the  Nobel  address  was  largely  focused  on 
the  work  that  we  were  doing  with  SVAO  mapping  and  so  on.   It  had 
nothing  to  do  with  cloning  or  recombinant  DNA.   It  was  done  in 
1980.   It  describes  the  dissection  of  SV40.   I  think  it's  called 
"The  Dissection  and  Reconstructions  of  Viral  Genomes".   It  has  to 
do  with  identifying  the  locations  of  the  genes  of  SVAO. 

Hughes:   I  have  the  paper.   [pause  while  Berg  scans  paper] 

Berg:    You  have  to  find  out  what  references  29  and  30  are  here.   Because 
I  say,  "Since  that  time  there  has  been  an  explosive  growth  in  the 
application  of  recombinant  DNA  methods  for  a  number  of  novel 
purposes  and  challenging  problems.   This  impressive  progress  owes 
much  of  its  impetus  to  the  growing  sophistication  about  the 
properties  and  use  of  restriction  endonucleases,  the  development 


'Paul  Berg,  "Dissections  and  reconstructions  of  genes  and 
chromosomes,"  Nobel  lecture,  December  8,  1980,  Bioscience  Reports  1981, 
1:269-287. 


118 

of  easier  ways  of  recombining  different  DNA  molecules,  and,  most 
importantly,  the  availability  of  plasmids  and  phages  that  made  it 
possible  to  propagate  and  amplify  recombinant  DNAs  in  a  variety  of 
microbial  hosts  (see  references  29  and  30  for  a  collection  of 
notable  examples.)" 

If  you  go  back  there,  my  guess  is  that  you  might  very  well 
find  that  these  are  references  to  either-- 

Hughes:   I'll  get  them  for  next  time.1 

Berg:    I  have  never  failed  to  acknowledge  Cohen  and  Boyer.   I  can  give 

you  copies  of  many  lectures  and  talks;  I  always  acknowledge  Cohen 
and  Boyer  for  being  the  key  people  in  developing  cloning 
technology.   And  there's  no  question  that  we  did  not  clone 
anything.   I  have  said  that  many  times.   The  confusion  lies  in 
whether  we  were  aiming  to  clone  things  and  didn't  understand  the 
system  well  enough  to  know  that  we  couldn't  have  cloned  things  the 
way  we  did  it.   But,  in  point  of  fact,  as  I've  told  you,  our  aim 
was  to  get  things  into  mammalian  cells,  not  to  clone  things  in 
bacteria.   That  was  something  that  somebody  raised  as  a 
possibility  from  the  point  of  view  of  risk,  which  was  never  in  our 
plans . 


More  on  Berg's  SVAO  Experiment:  No  Expectation  of  Cloning 


Hughes:   Well,  another  issue  was  raised:  if  you  had  known  that  the  way  the 
SVAO  DNA  was  inserted  would  prevent  replication,  would  you  have 
recommended  the  moratorium? 

Berg:     What  do  you  mean  by  if  we  had  known? 

Hughes:   Well,  from  what  I  understand  of  the  science,  the  way  the  DNA  was 
actually  inserted  interfered  with  a  gene  that  was  required  for 
replication. 

Berg:     Oh,  okay.   The  piece  of  bacterial  DNA  we  used  is  actually  a 

plasmid.   It  has  a  replication  origin  and  the  genes  needed  for 
replication,  and  it  has  these  three  bacterial  genes.   We  opened 
this  piece  of  DNA  in  a  region  of  the  sequence  which  is  necessary 


1  References  29  and  30  refer  to  issues  of  Science  devoted  to  articles 
on  recombinant  DNA;  there  are  no  papers  by  Cohen  and/or  Boyer.   (Science 
1977,  196,  no.  4286,  April  8,  1977;  1980,  209,  no.  4463,  September  19, 
1980.) 


119 


for  it  to  replicate.   And  so,  when  we  stuck  the  SV40  into  it,  this 
molecule  would  not  have  replicated  in  E.  coli.   We  didn't  know 
that.   But  since  that  wasn't  our  objective,  it  would  not  have  even 
occurred  to  us  to  know  whether  this  was  going  to  replicate  in  E. 
coli. 

Actually,  the  way  to  make  it  replicate  in  E.  coli  would  have 
been  to  make  a  dimer.   So  had  that  been  our  goal,  we  would  have 
solved  that  issue.   But,  that  wasn't  our  goal.  We  weren't 
thinking  along  those  lines.   Our  goal  was  to  open  SV40  in  a  region 
which  left  the  SV40  genes  intact.  We  could  have  used  random 
opening  of  SVAO  because  some  of  the  molecules  would  have  been  open 
in  a  region  which  did  not  inactivate  any  genes. 

It  turns  out  that  Rl  broke  right  into  one  of  the  SV40  genes. 
And  while  we  could  have  made  a  recombinant,  no  problem,  by  Rl,  it 
would  not  have  been  able  to  express  the  major  capsid  protein. 
Stan  has  raised  the  point,  "Aha,  you  would  never  have  been  able  to 
clone  it."   And  I  said  to  him,  "Stan,  that  was  never  our 
intention,  that  wasn't  the  goal  of  the  experiment.   The  goal  of 
the  experiment  was  to  create  a  transduction  system  for  mammalian 
cells.   We  weren't  looking  to  put  things  into  £.  coli."   I 
acknowledge  that  freely.   And,  therefore,  our  experiments  didn't 
open  the  door  to  cloning  in  bacteria  because  that  wasn't  what  we 
had  in  mind. 

All  that  we  did  was  raise  the  consciousness  that  you  could 
join  DNA  molecules  together  outside  a  cell.  And,  once  you  begin 
to  think  about  what  was  necessary  to  get  recombinant  DNAs  to  do 
certain  things,  people  began  being  more  sophisticated  about  what 
gets  joined  to  what  and  where  it  gets  joined. 


More  on  the  Biohazards  Controversy 


Berg:     The  Morrow  experiment,  if  I  remember  correctly,  would  have  been 
early  '74. 

Hughes:   And  the  first  Cohen-Boyer  paper  was  published  in  November  1973. 

Berg:     [June]  "73  was  the  Gordon  conference,  because  Boyer  reported  the 
first  cloning  experiment  which  got  a  lot  of  people  uptight.   And 
'  74  was  the  Morrow  experiment .   The  ' 73  experiment  reported  at  the 
Gordon  conference  got  people  excited  and  triggered  the  National 
Academy  of  Sciences  to  get  involved.   It  got  me  involved. 


120 


MIT  Meeting  to  Discuss  Biohazards,  1974 


Berg:    As  we  were  readying  the  meeting  at  MIT  to  discuss  how  to  advise 
the  Academy,  up  comes  the  Morrow  et  al.  paper.   The  Morrow  paper 
shows  that  anything  can  be  inserted  into  a  plasmid  and  be  cloned. 
That  raised  the  stakes.   Stan  Cohen  said,  "I'm  getting  telephone 
calls  from  all  over  the  world,  people  asking  me  to  send  them 
pSClOl.   What  should  I  do?"  So  when  I  went  to  MIT  for  the 
meeting,  it  was  with  the  background  that  it  was  now  clear  anything 
could  be  put  into  E.  coli. 

Some  of  the  things  that  Stan  Cohen  said  people  were  calling 
him  about  sounded  ominous!   Should  the  tetanus  toxin  gene  be  put 
into  E.  coli  and  enable  E.  coli  to  do  something  it  can't 
ordinarily  do?   So,  those  were  the  things  that  led  to  our 
consideration  of  a  moratorium.   Interestingly  enough,  Stan  was  not 
involved  in  that  committee  meeting  at  MIT,  nor  was  Boyer,  nor  was 
Hogness,  nor  was  Davis.   But,  as  soon  as  we  brought  back  the 
outline  of  what  we  had  decided  on  at  that  meeting,  they 
immediately  wanted  to  have  their  names  associated  with  it.   That's 
how  they  became  signatories  of  the  "Berg"  letter. 

Hughes:   Am  I  remembering  correctly  that  you  were  the  organizer  of  that  MIT 
meeting? 

Berg:     Yes. 

Hughes:   Had  you  invited  them? 

Berg:     No.   I  had  invited  primarily  people  who  had  been  involved  in 

Asilomar  I,  who  had  some  experience  in  thinking  about  the  risks  of 
this  kind  of  experimentation.   Jim  Watson,  because  he  had  been 
outraged  by  some  of  the  things  that  people  wanted  to  do  with 
viruses.   Dave  Baltimore,  because  he  clearly  had  been  one  of  the 
principals  in  working  with  animal  viruses,  particularly  tumor 
viruses.   So,  all  the  people  who  came  to  that  meeting  were  people 
who  had  been  involved  in  that  first  Asilomar  meeting. 


Berg:     I  also  invited  Maxine  Singer  but  she  couldn't  come.   I  don't 

remember  if  we  invited  anybody  else  that  couldn't  come.   But  we 
felt  that  seven  was  a  reasonable  size.   Remember,  we  were  not  on  a 
mission  to  do  what  we  did.   [laughter]   I  had  been  asked  by  the 
president  of  the  National  Academy  of  Sciences  to  advise  the 
academy  on  how  to  respond  to  the  concerns  that  had  been  voiced  at 
the  Gordon  conference.   It  was  a  very  limited  request.   I  just 
took  it  on  myself  to  say  that  I  wasn't  going  to  advise  the 


121 


president  of  the  academy  on  my  own.   And  what  I  would  do  is  try  to 
bring  some  people  together  that  I  thought  would  be  thoughtful  and 
do  something  about  it  and  just  asked  these  seven  or  eight  people. 
It  turned  out  there  were  six  others  beside  myself. 


The  "Berg"  Letter,  July  26,  19741 


Berg:    But  when  I  came  back  to  Stanford,  I  reported  that  the  group's 

decision  was  to  publish  a  letter  in  Science  and  Nature,  calling 
attention  to  the  potential  biohazards  and  offering  recommendations 
on  how  to  proceed.   Stan,  and  Herb,  and  Ron  Davis,  and  Dave 
Hogness  immediately  wanted  to  have  their  names  associated  with 
this. 

Hughes:   Why? 

Berg:     I  don't  know.   Stan  Cohen  and  Dave  Hogness  later  thought  the  whole 
thing  was  stupid.   Nevertheless,  they  signed  the  letter,  as  did 
Jim  Watson.   At  the  [Asilomar  II]  meeting,  Jim  Watson  was  most 
vociferous  about  calling  for  a  moratorium.   Why  they  did  it?   I 
don't  know.   Maybe  they  thought,  as  we  did,  that  there  was  enough 
uncertainty  that  it  was  a  reasonable  thing  to  do.   Maybe  they  did 
it  because  they  wanted  to  have  their  name  associated  with 
something  that  would  have  that  flavor.   I  just  don't  know.   But, 
we  did  not  exclude  them.   There  was  never  a  question  of  voting, 
should  we  or  should  we  not.   These  people  wanted  to  put  their  name 
to  it. 

My  recollection  is  that  I  probably  thought  it  was  a  good 
thing,  because  they  were  active  participates,  their  names  were 
very  well-connected  with  recombinant  DNA  already.   Therefore,  if 
their  names  were  on  it,  this  [letter]  would  have  more  of  an  impact 
on  the  rest  of  the  community. 

Hughes:   Did  they  come  to  you  and  ask  to  be  put  on  the  letter? 

Berg:    Yes.   Well,  I'm  sure  I  circulated  a  draft  of  what  we  had  come  up 
with.   And  they  said,  could  they  put  their  names  on  it  too? 

Hughes:   I  can  see  that  it  would  work  to  support  the  letter's 

recommendations  to  have  people  sign  this  letter  who  were  actually 
involved  in  these  experiments. 


1  Paul  Berg  and  ten  signatories,  "Potential  biohazards  of  recombinant 
DNA  molecules,"  Science  1974,  185:303  (July  26,  1974). 


122 

Berg:     I  think  that  was  in  the  equation.   There  was  never  any  thought 

that  they  should  be  excluded.   And  so,  I  don't  think  there  was  a 
big  argument,  for  example,  pros  and  cons.   It  seems  not  an 
unnatural  thing  and  might  very  well  have  been  obvious  that  if 
you've  got  the  very  people  who  were  most  involved  in  the  work, 
actual  participants  of  the  work,  the  letter  would  have  more  force, 

Don't  forget,  the  letter  was  addressed  to  colleagues  around 
the  world;  it  wasn't  addressed  to  the  local  community.   The  point 
is,  if  you  had  names  like  Cohen,  and  Boyer,  and  Hogness,  and 
Davis,  in  addition  to  the  other  people  that  had  been  on  it,  the 
letter  would  have  more  impact,  more  acceptance. 


The  Nobel  Prize  in  Chemistry,  1980 
Singling  Out  Berg 


Hughes:   Let's  go  on  to  a  discussion  of  the  Nobel  Prize,  even  though  it's  a 
few  years  forward  in  time.   I  read  the  article  in  the  November 
1980  of  Science  which  quotes  the  press  release  from  the  Swedish 
Royal  Academy:  "Berg  was  the  first  investigator  to  construct  a 
recombinant  DNA  molecule,  i.e.,  a  molecule  containing  parts  of  DNA 
from  different  species."1   Do  you  think  that  phraseology  is  meant 
to  give  the  reader  a  fully  descriptive  idea  of  what  the  research 
was  about?  Or  was  it  a  way  of  tacitly  explaining,  to  those  who 
really  understood  the  body  of  research,  why  you  had  been  singled 
out  rather  than  someone  else,  namely  that  you  were  the  only  one  to 
have  joined  DNA  from  different  sources? 

Berg:     I  don't  know  who  is  the  author  of  that  statement;  the  actual 

statement  for  the  Nobel  Prize  talks  about  my  early  work  as  well  as 
the  recombinant  DNA.   Not  as  explicitly  as  that.   Essentially,  we 
worked  in  nucleic  acids  and  recombinant  DNA.   I  don't  even  know 
where  the  statement  you  quoted  comes  from. 

If  you  talk  about  recombinant  DNA,  you  talk  about  joining 
two  different  kinds  of  DNA  molecules  together,  and  our  experiment 
was  clearly  the  first  to  do  that.   What  Peter  Lobban  did  was  make 
dimers.   What  Sgaramella  did  was  make  dimers.   And  whoever  this 
other  guy  was? 


1  G.B.  Kolata,  "The  1980  Nobel  Prize  in  Chemistry,"  Science  1980, 
210:887-889. 


123 


Hughes:   Jensen. 

Berg:     Jensen  made  dimers.   Is  that  a  distinction?   I  don't  know.   I 
disdain  the  kind  of  identif ication--"the  father  of  genetic 
engineering",  all  that  kind  of  hype  that  comes  around.   I  have 
never  claimed  that  we  developed  cloning;  we  didn't.   What  we  did 
was  develop  a  way  to  join  two  different  DNA  molecules  together 
outside  of  a  living  cell—period. 


Berg's  Opinion  of  His  Best  Work 


Hughes:   If  you  were  the  Nobel  committee  and  could  choose  amongst  your  own 
scientific  achievements,  would  you  choose  the  recombinant 
DNA  work? 

Berg:    You  mean,  would  I  have  chosen  that  bit  of  work  for  me  to  get  the 
prize? 

Hughes:   What  I'm  really  asking  is,  do  you  consider  it  to  be  your  best 
work? 

Berg:    No,  no,  no,  not  at  all.   But,  you  see,  that's  not  the  criterion 

that  the  Nobel  committee  uses.   The  man  who  shared  the  prize  with 
Kornberg,  [Severe]  Ochoa,  got  the  prize  for--presumably--having 
discovered  the  means  for  making  RNA.   But  in  fact  we  know  that  the 
enzyme  he  discovered  doesn't  make  RNA  at  all.   It's  like  that 
terminal  transf erase;  it's  a  dumb  polymerase  that  just  polymerizes 
nucleotides  at  random.   But  Ochoa  was  one  of  the  towering  figures 
in  biochemistry.   Everybody  would  have  said  that  this  guy  was 
going  to  get  the  Nobel  Prize.   What  did  he  get  it  for?   He  got  it 
for  something  which  he  would  not  have  identified  as  his  most 
notable  achievement.   I  think  you  can  say  that  for  any  number  of 
Nobel  awards. 

I  might  have  told  you  very  early  in  our  interview  process 
that  I  thought  the  most  significant  and  most  innovative  piece  of 
work  that  I  did  was  as  a  postdoc  when  I  joined  Kornberg 's  lab,  in 
the  discovery  of  this  new  class  of  compounds  for  activating 
molecules  for  assembly  into  larger  molecules.   I  think  that  was 
much  more  creative. 


124 


Speculations  on  Why  Berg  Was  Chosen 

Berg:     But  remember,  recombinant  DNA  had  two  things  going  for  it.   One 
was  it  was  at  the  focus  of  a  public  policy  debate  all  over  the 
world,  in  Sweden  as  well.   And  two,  it  had  an  enormous  impact. 
What  the  Nobel  Prize  often  rewards  is  the  way  science  is  changed 
as  a  consequence  of  that  discovery. 

Carl  Cori  was  one  of  the  great  figures  of  biochemistry  in 
the  1940s.   He  and  his  wife  Gerty  shared  the  Nobel  Prize  for 
discovering  an  enzyme  that  breaks  down  glycogen.   If  you  ask  any 
student  today  who  Carl  Cori  was,  they  wouldn't  know.   And  two,  if 
you  asked  them  if  they  thought  that  glycogen  phosphorylase  had  a 
major  impact  or  changed  the  face  of  biochemistry,  they  would 
answer  no.   But,  in  its  day,  it  was  an  innovative  finding. 
Remember,  they  had  already  established  themselves  as  premiere 
biochemists . 

So,  I'd  like  to  think  that  the  Nobel  Committee  also 
considered  the  highly  regarded  work  that  I  had  done  long  before 
the  recombinant  DNA.   Perhaps  some  people  were  supportive  because 
I  was  a  leader  in  the  public  policy  part  of  it.   And  in  some 
circles  that  seems  to  be  more  important  than  the  scientific  part. 

Hughes:   Really? 

Berg:    Yes.   I  don't  know  whether  the  people  on  the  Nobel  committee  used 
that.   More  likely,  however,  is  the  enormous  impact  and  the  way 
[recombinant  DNA  technology]  changed  the  way  biology  is  done. 
Anybody,  looking  back,  could  ask  how  this  enormous  new  kind  of 
science  began.   Where  does  it  go  back  to?   It  starts  with  the 
demonstration  that  you  can  join  DNA  molecules  together.   And  then 
a  lot  follows  from  that. 

I  was  invited  shortly  after  the  public  policy  issue  broke- 
publication  of  the  moratorium  letter—to  come  and  give  a  lecture 
in  Sweden,  which  the  king  attended.   It  was  a  big  public  lecture. 
Swedish  opinion  was  much  more  interested  in  the  issues  of 
responsibility  in  science,  and  ethics,  and  safety.  And  so,  I 
could  have  become  a  hero,  I  don't  know.   There  might  have  been 
some  people  that  would  have  regarded  what  I  did  as  being 
courageous.   I  don't  know.   I  didn't  think  it  was  courageous. 

But,  people  look  at  that  whole  episode  in  different  ways. 
It  has  really  been  amazing.   We  focused  on  it  very  narrowly:  is 
[recombinant  DNA  research]  safe,  is  it  not  safe?  People  saw  it  as 
raising  a  big  ethical  debate  and  us  displaying  an  enormous  level 
of  ethical  concerns.   But  we  saw  it  as  a  public  health  issue. 


125 


Hughes:   Well,  you  have  been  criticized  for  narrowing  the  debate  to  the 
issue  of  biohazards. 

Berg:    That's  right.   And,  I  think  that  diminishes  any  attribution  of 
high  ethical  standard.   If  you  said  we  were  in  this  to  really 
raise  the  consciousness  of  the  world  about  the  impending  change 
and  the  way  things  were  going  to  be  done,  we  didn't  do  that! 

Hughes:   I  think  associated  in  the  public  mind  is  the  idea  of  scientists 
policing  themselves. 

Berg:     Yes. 

Hughes:   That  idea  could  have  influenced  the  Nobel  committee. 

Berg:    After  the  award  was  announced,  there  was  an  editorial  in  Wature, 
or  a  little  blurb  about  the  award.   My  recollection  is  that  the 
guy  who  wrote  it  certainly  acknowledged  that  idea  could  easily 
have  had  an  impact  on  the  choice  of  Berg.   I  mean,  nobody  tried  to 
say,  you  didn't  deserve  it  because  the  science  you  did  was  trivial 
or  whatever.   Without  being  too  modest,  people  recognized  the 
science  that  I  had  done  was  first  class.   The  recombinant  DNA 
thing  is  a  thing  with  a  big  impact.   It's  easy  to  latch  on  to  that 
and  say  that's  what  you  give  the  prize  to.   I  don't  know  if  I 
would  have  gotten  the  prize  for  the  previous  work  alone. 

So,  if  you  add  it  up,  there's  a  first-class  science 
background,  there's  some  kind  of  association  with  a  very  important 
scientific  breakthrough,  and  then  a  leadership  role  in  trying  to 
manage  the  impact  of  that  breakthrough.  When  it's  taken  together, 
people  say,  well,  okay.   There's  another  point  as  well.   The  Nobel 
prize  is  limited  to  three  individuals. 

Hughes:   And,  they  have  to  be  living. 

Berg:    They  have  to  be  living.   There  have  been  a  number  of  instances 

where  people  were  passed  over  for  the  prize  because  it  would  have 
exceeded  three.   In  other  words,  there  were  four  people  that 
contributed  equally,  or  this  one  person  was  unequivocally 
associated,  and  there  are  three  others,  and  these  three  don't  get 
it.   In  fact,  such  a  thing  actually  happened.   Phil  Sharp  and  one 
other  person,  Richard  Roberts,  got  the  prize  for  discovering 
introns.   Now,  the  person  at  Cold  Spring  Harbor  who  actually 
discovered  the  anomaly  that  led  to  the  insight  [Tom  Broker]  didn't 
get  it.   And  when  people  talked  about  why  he  didn't  get  it,  it  was 
explained  that  it  was  because  he  and  his  wife  had  collaborated  on 
it  and  that  would  have  made  four  people. 

Hughes:   That's  tough. 


126 


Berg:     Okay,  so  now  the  question  is,  why  didn't  Cohen  and  Boyer  get  it 
instead  of  me? 

Hughes:   Or,  why  didn't  Berg,  Cohen,  and  Boyer  get  it? 

Berg:    Because,  the  Nobel  Committee  believed,  I  think,  that  the  critical 
and  important  contributions  to  recombinant  DNA  were  cloning  and 
sequencing.   Cloning  without  sequencing  is  trivial.   Sequencing 
without  having  specific  cloned  pieces  of  DNA  to  sequence  is  also 
useless.   Developing  both  were  technical  feats,  and  the  two 
together  are  what  really  made  the  molecular  genetics  revolution. 
People  cloned  DNA  and  sequenced  it.   Okay?   So,  why  not  Cohen  and 
Boyer,  and  Gilbert  and  Sanger?  That's  four.   So,  you  could  say, 
one  compromise  is  not  Cohen  and  Boyer,  but  Berg.   It's  difficult 
to  know  what  the  thinking  of  the  Nobel  Committee  was. 

Hughes:   In  speculating  how  the  biohazards  controversy  may  have  elevated 

you  in  the  Swedish  consciousness,  I  wonder  if  Cohen's  and  Boyer 's 
commercial  ventures  worked  to  their  disfavor  in  the  eyes  of  the 
Nobel  committee? 

Berg:    I  don't  know.   The  secrecy  that  shrouds  the  decisions  about  the 
Nobel  are  remarkably  well  kept.   The  Swedes  have  managed  to  keep 
that  process  unpolluted  by  leaks.   There  may  be  speculations,  and 
there  are  people  who  try  to  hype  themselves  or  somebody  else  by 
saying  they're  being  considered,  etc.   But,  what  I  know  from  my 
Swedish  friends  who  are  involved  in  the  process,  it  is  very 
tightly  guarded. 

If  somebody  were  to  suggest  to  them  that  there  were 
extraneous  issues  that  were  considered  or  played  a  role,  they 
would  deny  it  vehemently.   People  involved  tell  me  they  spend  the 
entire  summer  researching  a  few  candidates,  going  back  through 
every  paper,  every  collateral  paper;  trying  to  document  priority 
is  very,  very  important  to  them.   In  the  end,  this  whole  process 
winnows  down  to  a  few  names,  and  then  it's  the  respective  Nobel 
academies  that  vote  on  them. 

Recommendations  from  the  individual  committees  have  been 
reversed;  that  is,  committees  have  come  in  with  a  recommendation, 
and  the  whole  academy  has  rejected  it  and  selected  another 
candidate  from  the  small  group.   So,  who  knows?  Yes,  in  the  final 
vote  of  the  academy,  it's  quite  conceivable  that,  without  anybody 
conceding  it,  sentiment  plays  a  role,  because  they  all  have  to 
vote.   And,  if  some  guy  says  I  admire  this  person  for  what  he 
stood  up  for  and  what  he  did,  it  could  sway  their  vote.   But,  I 
don't  think  those  issues  would  have  any  impact  at  all  with  the 
screening  and  review  committees. 


127 
Influences  on  the  Choice  of  the  Nobel  Award 


Hughes:   I  thought  of  Gary  Mullis  when  we  were  discussing  the  fact  that  the 
prize  is  usually  given  for  a  body  of  research. 

Berg:    In  that  case,  he  had  done  nothing  before. 
Hughes:   So  there  are  exceptions  to  the  rule. 

Berg:    Michael  Smith,  who  was  the  other  guy  who  got  the  prize  that  year, 
made  one  contribution  when  he  was  a  postdoc  of  Rhorana's.   He 
hadn't  ever  distinguished  himself  in  other  ways;  I've  known  him 
for  a  long  time.   The  idea  of  being  able  to  make  targeted 
mutations  in  DNA  the  way  he  did  it  certainly  has  had  an  enormous 
impact.   And,  my  guess  is,  that's  why  he  got  it.   If  you  ask,  was 
it  a  great  intellectual  breakthrough,  a  great  intellectual 
inspiration,  the  answer  is  no. 

Hughes:   Why  did  the  most  preeminent  prize  in  science  go  to  two 
technological  achievements? 

Berg:    I  don't  know  which  ones  you're  referring  to. 
Hughes:   I'm  referring  to  PCR  and  mutagenesis. 

Berg:     People  have  speculated,  and  I  have  no  insight  at  all,  as  to 

whether  there  are  contending  forces  on  the  Nobel  Prize  decision. 
Some  would  like  to  make  it  more  practical;  some  favor  more 
theoretical.   Some  favor  more  basic,  others  more  applied.   It 
almost  seems  to  alternate.   Like  one  year  it's  a  group  of 
physicists  who  developed  a  machine,  another  one  it's  a  cosmologist 
who  has  come  up  with  a  grand  theory. 

And  the  same  thing  has  happened  in  biology.   When  I  say 
basic,  it  could  be  the  crystallization  and  determination  of  the 
structure  of  a  protein.  And  then  the  next  year,  it's  somebody  who 
has  developed  a  whole  series  of  compounds  that  are  promising  for 
cancer  chemotherapy.   Those  two  are  very  different  kinds  of 
intellectual  activities.   I  don't  know  whether  within  the  academy 
they  debate  this  kind  of  issue. 

Nobel's  objective  was  to  reward  advances  in  science  that  had 
an  impact  on  society,  and  he  suggested  that  it  be  for  a  discovery 
in  the  year  of  the  award.  Well,  very  few  of  the  prizes  have  been 
given  for  things  that  have  had  impact  in  that  year,  because, 
rightfully  so,  some  period  of  time  is  needed  to  see  whether  the 
discovery  is  just  a  flash  or  whether  it  has  some  lasting  effect. 
And  so  you  have  to  wait. 


128 

Yet  More  on  Recombinant  DNA 
[Interview  A:  November  5,  1997)  ft 

Terminal  Transferase 


Berg:    The  existence  of  terminal  transf erase  was  long  known.   It  was  a 
curiosity,  because  it  was  the  first  enzyme  that  actually 
synthesized  DNA  without  a  template.   That's  not  quite  true. 
Kornberg  had  shown  that  DNA  polymerase  would  synthesize  some  kind 
of  funny  polymer  without  a  parent  template.   But,  in  this  one 
terminal  transferase,  you  needed  a  primer.   You  needed  DNA,  and 
all  it  did  was  add  nucleotides  on  to  the  ends. 

My  recollection  is  that  in  Bollum's  work  it  wasn't  known  if 
transferase  would  add  on  to  the  appropriate  3-prime  end  of  a 
double-stranded  DNA  if  it  was  blunt.   It  had  to  be  a  protruding 
primer  end;  that's  what  Peter  Lobban  essentially  discovered  and 
quickly  related  to  us. 


Creating  Permuted  Linear  Molecules  with  "Tails" 


Berg:    In  fact,  I  thought  a  little  bit  about  it  after  our  conversation. 
Our  original  plan  was  to  open  the  SVAO  circle  at  random  sites, 
because  we  knew  we  were  going  to  put  tails  on  the  ends.   So,  it 
didn't  make  any  difference  where  we  opened  it.   And  by  cleaving 
the  DNA  at  random  sites,  we  would  have  a  population  of  linear 
molecules  that  were  permuted,  that  is,  they  would  have  different 
ends  because  they  were  opened  at  different  places.   That  would 
minimize  the  risk  that  where  we  were  actually  making  the 
attachment  to  the  other  molecule  would  interfere  with  some 
essential  function  in  the  SV40. 

So,  we  spent  a  lot  of  time  using  an  enzyme  called  pancreatic 
DNase,  DNase-1.   It  had  been  discovered  by  others,  and  we 
concurred  that  if  you  do  it  in  the  presence  of  manganese,  you  make 
primarily  a  single  cut,  and  then  it  stops.   So,  we  were  cutting 
with  pancreatic  DNase,  making  linear  molecules  with  different 
ends,  polymerizing  A's  and  T's  on  to  the  end,  and  then  joining 
them  together.   But  when  John  Morrow  discovered  that  EcoRl  made  a 
unique  cut,  we  didn't  know  exactly  which  gene  the  cleavage  was  in. 
It  turned  out  it  was  in  the  gene  that  specifies  the  capsid 
protein. 


129 


If  I  remember  correctly,  we  used  the  Rl-cut  linears,  because 
they  were  always  uniform.   And  because  they  were  uniform,  we  knew 
that  they  had  5-prime  protruding  ends,  and  that's  not  the  kind  of 
thing  that  you  want  this  terminal  transf erase  to  be  adding  to.   We 
needed  a  way  to  remove  the  5-prime  extensions  to  leave  3-prime 
extensions,  and  that  was  done  with  an  enzyme  which  had  been 
discovered  at  Stanford,  called  exonuclease-7 .   That  enzyme 
specifically  degrades  double-stranded  DNA  starting  at  the  5-prime 
end  and  going  in,  therefore  leaving  3-prime  extensions.   So  that 
was  essentially  the  strategy  and  how  we  evolved  to  it. 


Choosing  the  Best  "Tails' 


Berg:    The  only  other  thing  that  I  forgot  to  mention  was,  we  spent  a  lot 
of  time  trying  to  figure  out  which  were  the  best  tails  to  use.   We 
originally  started  out  to  ask  whether  G-C  tails  were  the  best  to 
use.   It  didn't  make  any  difference  in  principle  which  you  used  if 
you  wanted  so-called  cohesive  ends.   But  it  turns  out  it  was  very, 
very  difficult  to  polymerize  G's  onto  the  ends  of  DNA.   There  was 
a  standing  joke  in  the  department  that  if  you're  going  to  have  any 
trouble  with  anything  in  DNA,  it's  going  to  be  with  G.   People 
tried  to  make  dGTP  and  that  always  failed  and  the  others  worked 
well,  and  so  on  and  so  forth. 

The  reason  why  we  were  having  trouble  is  G  forms  internal 
polymers  with  itself.   And  so  if  you  polymerize  onto  the  end  and 
make  a  long  tail  of  G,  the  chances  are  it  is  going  to  form  some 
kind  of  funny  structures.   In  fact,  G-C  forms  triple-stranded 
structures.   When  that  was  discovered,  we  realized  using  G-C  was 
causing  bad  things.   So  we  went  back  to  using  A-T.   I  don't  think 
we  ever  published  the  ins  and  outs  and  failures  because  we 
published  in  PNAS ,  and  PNAS  has  a  limited  amount  of  space.   We 
went  right  to  the  direct  thing  [A-T].   Actually,  Bob  Symons,  who 
was  an  Australian  visitor,  did  a  lot  of  the  work  trying  to  produce 
these  G-C  tails. 


The  Jensen  et  al.  Paper  and  Biochemical  and  Biophysical  Research 
Commun ications 


Hughes:   Have  you  said  all  you  care  to  say  about  the  Jensen  paper? 
Berg:    Other  than  I  didn't  know  it  existed.   I  never  heard  of  it. 


130 

Hughes:   What  about  the  journal  itself? 

Berg:    BBRC  [Biochemical  and  Biophysical  Research  Communications]! 

Hughes:   Yes. 

Berg:    I  was  an  editor  of  it  for  a  while  in  its  earliest  days.   I  know  I 
wasn't  an  editor  at  that  point.  Although,  my  memory--.   It 
started  out  as  a  journal  presumably  to  allow  short  hot  new 
findings  to  be  published.   It  certainly  did  that  for  some  things. 
But  after  a  while,  it  accumulated  a  lot  of  junk.   I  mean  just 
things  that  people  might  have  identified  as  critically  hot  papers 
the  way  they're  referred  to  today.   I  went  off  it  [as  editor] 
because  it  was  getting  to  be  a  nuisance.   You  see,  the  articles 
are  quite  short  because  of  limited  space.   They  were  reproduced 
from  your  submitted  text,  thereby  circumventing  the  need  for 
redactory  and  all  of  that  business.   It  was  intended  as  quick 
publication;  quick  because  they  would  just  photocopy  what  you  sent 
them.   It  never  had  a  very  good  reputation.   It  was  viewed  as  "bio 
quickies".   I  just  decided  I  didn't  want  to  be  on  it  anymore.   I 
don't  remember  exactly  when  it  started. 

[Berg  indicates  Jensen  paper]  This  is  volume  43,  1971,  so 
you  can  see  it  must  have  started  way  back  in  the  sixties.   It  came 
out  every  month,  and  it  was  just  a  nuisance.   I  don't  think  I  ever 
read  the  bio  quickies  after  I  left  the  editorial  board.   I  mean  I 
rarely  look  at  it.   But  people  do  read  it,  do  refer  to  it, 
particularly  people  who  are  worried  about  somebody  scooping  them. 
There's  a  journal  that  publishes  just  the  table  of  contents  of 
various  journals.   So,  people  have  resorted  to  essentially 
skimming  the  titles.   I  don't  remember  ever  seeing  the  Jensen 
paper. 

Hughes:   Does  the  fact  that  Jensen  et  al.  chose  to  publish  in  this  journal 
suggest  that  they  knew  that  they  had  something  hot?   You 
characterized  the  journal  as  a  place  for  hot  papers. 

Berg:    No.   It  had  two  features,  one  is  you  could  submit  short 

communications,  and  they  were  published  quickly.   And,  they  were 
almost  unreviewed. 

Hughes:   The  summary  to  their  paper  doesn't  mention  commercial 

applications.   I  wonder  if  they  knew  the  significance  of  what  they 
had  done? 

Berg:    Actually,  the  last  sentence  is  interesting.   "Polynucleotide 

ligase"--DNA  ligase--"does  not  covalently  join  the  single  strands 
of  these  synthetic  catenanes."   In  other  words,  if  you  put  A's  and 
T's  on  the  ends,  and  then  they  join,  there  are  going  to  be  gaps 


131 


because  there's  no  way  to  control  how  long  the  tails  are.   So  you 
can  have  twenty- five  on  this  one  and  ten  on  this  one,  and  they'll 
form  ten  base  pairs,  but  then  there's  a  big  gap.   He  obviously 
didn't  appreciate  that  since  he  couldn't  control  the  length  of  the 
homopolymer  tails.   That's  why  we  and  Peter  ended  up  realizing 
that  when  you  hybridize  the  two  and  they  join,  DNA  polymerase  and 
the  four  triphosphates  can  be  used  to  fill  in  the  gaps  and  ligase 
to  seal  them. 

Hughes:   Jensen  hadn't  done  that? 

Berg:    I  haven't  read  the  paper,  but  in  the  last  sentence,  it  seems  to 
indicate  that  he's  unaware  that  there  are  gaps,  which  would  be 
amazing.   Oh,  it  says,  "The  catenanes  formed  by  mixing  T7  DNA 
which  contained  synthetic  homologous  ends  were  apparently  not  as 
well  formed  as  those  which  occur  in  nature...   We  have  been  unable 
to  catalyze  covalent  joining  of  our  T7  DNA  catenanes  using  £.  coli 
polynucleotide  ligase,  even  though  numerous  experimental 
conditions  were  used  including  variations  in  temperature,  salt 
concentration  and  enzyme-substrate  level."   It  never  mentions  the 
fact  that  the  ends  are  unlikely  to  have  been  perfectly  matched. 

"Catenanes  of  T7  DNA  molecules  with  various  lengths  of 
homopolymeric  ends  were  also  not  joinable."  Last  paragraph: 
"Ligase  joining  during  concurrent  repair  DNA  synthesis  was  also 
attempted  under  conditions  identical  with  those  described  by 
Goulian  and  Kornberg."   But,  these  were  unsuccessful. 

This  was  submitted  March  12,  1971.   I  don't  have  my 
notebooks  to  know  where  that  stood  in  terms  of  our  work. 

Hughes:   I  don't  remember  when  the  Jackson,  Symons,  Berg  paper  was 
submitted. ' 

Berg:     [Berg  scans  publications  for  submittal  dates]   These  are  all 
November  '72. 


July  31,  1972. 


132 


The  Stanford  Biochemistry  Department's  Industrial  Affiliates 
Program 


The  Chemistry's  Industrial  Affiliates  Program  [IAP] 

Hughes:   What  I  really  want  to  talk  about  today  is  the  commercialization  of 
the  science.   I  thought  that  the  place  to  start  is  with  the 
biochemistry  department's  Industrial  Affiliates  Program.   Does 
that  seem  appropriate  to  you? 

Berg:     Yes. 

Hughes:   In  1970,  you  wrote  to  Carl  Djerassi  saying  that  you  were  "taken", 
that  was  your  word,  by  the  brochure  he  had  sent  to  you  on  the 
Industrial  Affiliates  Program  in  the  Department  of  Chemistry,  and 
that  you  were  interested  in  starting  something  similar  in  the 
Department  of  Biochemistry.1  Well,  it  apparently  took  almost  ten 
years,  because  the  first  time  any  such  idea  is  mentioned  in  the 
faculty  minutes  is  in  1979. 2  The  program  didn't  get  off  the 
ground  until  1980.   Was  there  a  reason  that  it  took  so  long  to  get 
going? 

Berg:    I  had  just  become  chairman  of  the  department  in  1969.   So  in  1970, 
if  that's  the  date  of  that  letter,  I  was  charged  with  the 
responsibility  of  helping  develop  resources  for  the  department. 
It  struck  me  that  the  chemistry  department  had  hit  on  a  mechanism 
which  was  really  very  interesting  because  it  generated  a  lot  of 
money . 

You  realize  that  in  1970  our  department  did  not  have 
something  it  could  go  out  and  sell  as  easily  as  it  did  in  1979. 
We  were  a  good  biochemistry  department.   We  were  very  highly 
thought  of.   Maybe  many  people  would  have  thought  we  were  number 
one.   But  the  question  is,  what  did  we  have  to  offer  to  industry? 

By  contrast,  chemistry  is  cranking  out  graduates  who  go  into 
the  chemical  industry,  and  more  likely  the  chemistry  department  is 
doing  research  that's  much  more  relevant  to  the  chemical  industry 
and  the  pharmaceutical  industry.   Carl  Djerassi  is  a  terrific 


1  Berg  to  Djerassi,  May  25,  1970  (Berg  papers,  Green  Library, 
Stanford,  S358,  box  2,  folder:  1970  A-H) . 

2  Minutes,  faculty  meeting,  Department  of  Biochemistry,  October  17, 
1979  (Arthur  Kornberg  papers,  SC  359,  Green  Library,  Stanford,  box  5, 
folder  1979).) 


133 


salesman,  and  given  his  connection  with  the  pharmaceutical 
industry,  he  was  able  to  actually  organize  at  one  point  forty 
companies  at  $25,000  per.   So  [Chemistry  was]  pulling  in  a  lot  of 
money.   We  didn't  have  anything  like  that,  but  we  were  well 
funded.   Part  of  the  resistance  in  the  biochemistry  department 
was,  who  needs  an  IAP! 


Broaching  the  Idea  of  a  Biochemistry  IAP 


Hughes:   Do  you  remember  that  you  broached  it  to  the  department  after  the 
interaction  with  Djerassi? 

Berg:     Oh,  yes,  I'm  sure  I  did. 

Hughes:   If  I  remember  that  letter  correctly,  one  of  your  arguments  is  that 
the  federal  funding  of  science  is  dropping  off.   Industry  has 
benefitted  all  along  from  academic  science;  it's  time  for  it  to 
pick  up  some  of  the  financial  burden. 

Berg:    That's  right.   That's  essentially  what  my  interpretation  was. 
[Berg  skims  copies  of  archival  documents  which  Hughes  has 
collected.]   Okay,  it  gets  picked  up  by  Arthur  Kornberg  in  1979. ' 
So,  the  only  thing  you  have  is  my  letter  to  Djerassi  that's  as 
early  as  1970. 

Hughes:   That's  right. 

Berg:    Also,  there's  a  '67  letter  in  which  Kornberg  planned  to  visit  at 
du  Pont,  and  they  wrote  to  him  about  that.2  I  remember  Arthur 
coming  back  and  just  being  totally  disillusioned  by  the  attitude 
of  industry,  and  the  way  they  did  research  and  so  on.   Arthur 
didn't  know  anything  about  the  affiliate  program  in  the  chemistry 
department. 


1  Memo,  Arthur  Kornberg  to  Dale  Kaiser,  September  10,  1979  (Kornberg 
papers,  SC359,  Green  Library,  Stanford,  box  5,  folder  1979). 

2  Burt  C.  Pratt  to  Arthur  Kornberg,  October  11,  1967  (Kornberg  papers, 
SC  359,  Green  Library,  Stanford,  box  26,  folder:  1967  A-L). 


134 
Stanford  Relationships  with  Industry 


Berg:    I  learned  at  that  time  that  Stanford  had  forty  Industrial 
Affiliates  Programs  going  in  the  university. 

Hughes:   In  1970? 

Berg:    I  think  so.   Stanford  was  really  a  very  entrepreneurial  place. 

The  aeronautical  and  engineering  departments  all  had  these  ongoing 
relationships  with  companies,  and  that,  I  think,  reflected 
Stanford's  close  relationship  with  industry,  which  led  to  Silicon 
Valley.   The  faculty  were  all  involved,  so  departments  nurtured 
these  kinds  of  relationships,  and  they  were  allowed  to  blossom 
unfettered.   If  somebody  was  entrepreneurial  enough  to  go  out  and 
create  a  program,  nobody  kept  tabs  on  them  as  far  as  I  know. 

My  recollection  doesn't  allow  me  to  tell  you  that  I  brought 
it  up  to  the  department.  The  letter  actually  mentions  something 
about  a  decrease  in  federal  funding.  I  remember  we  had  a  crisis 
during  my  chairmanship  when  there  was  a  threat  that  the  training 
grants  were  going  to  be  dropped. 

Hughes:   Right,  I  remember  seeing  a  reference  to  that. 

Berg:     And  the  training  grants  were  vital  to  our  program.   There  was  this 
threat  that  Congress  was  going  to  do  away  with  training  grants.   I 
went  to  Washington  and  lobbied,  and  so  on  and  so  forth.   And, 
probably  it  was  in  response  to  that  that  we  were  looking  for  new 
sources  of  funding. 

Hughes:   Which  you  didn't  come  up  with? 
Berg:     I  don't  know  the  dates  well  enough. 


Program  Project  Grant,  Institute  for  Research  on  Aging 


Berg:    But,  one  of  the  things  that  the  department  landed  was  a  program 
project  grant  from  the  Institute  for  Research  on  Aging.   The 
department  banded  together  four,  five,  six  people,  and  we  then 
made  a  proposal  to  the  aging  institute,  and  they  funded  us.    It 
only  terminated  about  two  years  ago.  We  held  the  grant  for  about 
four  cycles  of  five  years  each.   So  we  may  have  well  have  gotten 
it  during  this  period.  We  have  notes  in  the  department;  you  could 
find  out  when  we  started  the  aging  grant .   The  aging  grant  brought 
in  a  fair  amount  of  money. 


135 


But  what  happened  is,  the  politics  changed,  and  Congress  did 
not  disband  training  grants,  so  we  never  lost  the  training  grant. 
So  it's  conceivable  that  the  reason  nothing  ever  happened  out  of 
this  first  notion  about  the  affiliates  program  was  that  the 
department  was  well  funded.   If  the  training  grant  was  secure,  my 
guess  is  we  would  not  have  developed  sufficient  enthusiasm  for 
doing  the  affiliates  program. 


Sentiment  Against  lAPs 


Hughes:   But  it  wasn't  opposed  because  of  any  feeling  in  the  department 
that  an  affiliates  program  wasn't  appropriate? 

Berg:     There  were  those  feelings;  there  were  some.   I  know  that  after  we 
started  our  program,  and  I  went  around  the  country,  and  when  I'd 
be  visiting  at  Yale  or  at  Cambridge,  I  mentioned  the  affiliates 
program,  and  some  people  said,  That's  not  a  thing  that  academics 
do. 

I  always  justified  it  that  we  were  doing  was  what  we 
normally  do  here:  we  teach.   We  weren't  making  proprietary 
contracts  with  our  affiliates.   We  weren't  giving  them  first 
access  to  any  discoveries.   All  we  were  really  doing  was  providing 
expertise  that  was  keeping  them  abreast  of  what  was  happening  in 
their  field.   And,  I  saw  that  as  an  educational  mission.   We  were 
just  getting  our  tuition  in  different  ways;  we  were  getting  it 
through  companies. 


Functions  of  the  Biochemistry  IAP 


Hughes:   I  remember  seeing  in  one  of  these  documents  that  the  point  of  the 
program  was  not  to  provide  specific  information  for  specific 
industrial  programs,  but  to  provide  general  knowledge,  as  you're 
saying,  in  this  field  in  which  you  were  expert. 

Berg:     That's  right.   It  gave  companies  access  to  faculty.   That  is,  the 
original  thing  was  they  could  send  somebody  to  visit  the 
department  each  year  to  meet  with  faculty  and  students  and 
postdocs.   We  would  arrange  for  them  to  have  sessions  with 
students  and  postdocs  for  the  purposes  of  their  attempts  to 
recruit,  and  we  would  send  them  preprints  of  our  papers  before 
they  were  published,  and  they  would  get  these  quarterly.   And,  at 
the  end  of  the  year,  they  would  get  a  bound  volume  of  all  the 


136 


departmental  papers.   They  had  no  proprietary  rights  to  any 
discoveries,  materials,  or  inventions,  or  anything  like  that,  and 
that  was  it. 

Once  we  mounted  the  program,  everybody  in  the  department  was 
really  strongly  committed  to  it.   Everybody  agreed  that  they  would 
go  and  visit.   Furthermore,  each  company  would  have  a  visit  from 
somebody  in  the  department  who  would  spend  the  day  talking  to 
scientists  and  give  a  seminar  on  their  own  research,  and  so  on. 

I  dubbed  this  the  Friends  of  Biochemistry,  because  that's 
the  kind  of  affiliation  I  was  hoping  to  sell  and  also  would  expect 
to  have,  namely,  we're  going  to  help  you;  you're  going  to  help  us. 
The  notion  of  obligation  wasn't  ever  really  broached  as  a  selling 
point  because  I  think  most  of  these  companies  eschew  that  kind  of 
notion.   They  say,  "We  pay  our  taxes,  and  that's  funding  you 
guys."  But,  the  idea  was  that  it  was  mutually  beneficial.   They 
would  get  information.   And,  the  reason  we  were  able  to  sell  it 
was,  by  1979  Stanford  biochemistry  was  one  of  the  leading  groups 
in  genetic  engineering. 

Hughes:   Right,  and  that's  what  they  wanted. 

Berg:    And  that's  what  they  wanted.   Many  of  these  companies  were  totally 
in  the  dark  about  the  developments.   They  were  really  grasping  at 
straws,  and  that's  why  there  was  momentum  for  small  companies. 
Guys  who  had  ideas  about  what  they  could  do  with  recombinant  DNA 
essentially  didn't  go  to  big  companies,  because  [the  companies] 
didn't  understand  it. 

Hughes:   Were  companies  in  the  affiliates  program  less  likely  to  start 

there  own  recombinant  DNA  programs  because  they  had  a  relationship 
with  you?   Or  was  it  an  incentive? 

Berg:    It  was  more  likely  an  incentive.   The  invitation  to  our  affiliates 
to  attend  our  retreats  where  they  actually  heard  about  the  ongoing 
research  didn't  come  until  later.   So,  I  can't  cite  that  as 
something  that  would  have  spurred  them  on.   Because  if  they  had 
attended  our  retreats,  they  would  have  seen  that  all  kinds  of 
exciting  things  were  happening  which  they  were  not  part  of. 

Hughes:   You're  talking  about  the  departmental  retreats  at  Asilomar? 

Berg:    Yes,  that  came  later. 

Hughes:   Yes,  I  have  an  agenda  somewhere. 


137 


Launching  Biochemistry's  IAP  in  the  Late  1970s 


Berg:    We  began  to  discuss  an  affiliates  program  in  '78  or  '79.   By  that 
time,  I  think  Bob  Lehman  was  just  coming  to  the  end  of  his  five 
years  as  chairman.   We  were  doing  five-year  terms.   I  stepped  down 
in  '74.   Bob  Lehman  would  have  done  '74  to  '79.   So  I  suspect  it 
was  right  at  the  very  end  of  his  tenure  that  we  began  as  a 
department  to  become  enthusiastic  about  doing  this.   And  then  Dale 
Kaiser  became  chairman.   And  so  it  was  during  his  tenure  that  the 
program  actually  got  underway. 

Hughes:   Why  had  members  of  the  department  become  enthusiastic? 

Berg:    Well,  probably  two  things.   One  is,  given  the  notoriety  of  our 
department,  we  were  now  more  confident  that  we  had  something  to 
sell.   Before,  people  were  less  confident,  other  than  our  general 
renown,  that  we  had  anything  to  offer  to  commercial  companies. 

But,  when  it  became  clear  that  we  were  quite  out  front  in 
the  area  of  genetic  engineering,  we  felt  we  really  had  something 
to  sell,  and  it  would  be  negligent  perhaps  to  not  take  advantage 
of  that.   We  all  made  that  same  argument;  we  really  could  use  the 
money.   I  think  we  may  have  brought  up  what  Chemistry  was  getting 
out  it.   And  having  free,  undesignated  money;  most  grants 
designated  how  the  money  was  to  be  spent.  We  didn't  have  a  bank 
of  free  money.   And  so  here  was  a  way  to  accumulate  money  that 
would  guarantee  security  of  the  department. 

I  think  everybody  bought  into  it.   I  don't  remember  that 
there  was  anybody  who  was  opposed  to  it.  And,  as  I  say,  we  all 
committed  ourselves  to  that  list  of  offerings  that  we  were 
prepared  to  do,  and  that  meant  [each  faculty  member]  making  an 
effort  to  go  visit  one  of  the  companies.   Most  people  saw  that  it 
was  not  a  big  deal  because  they  were  traveling  a  lot.   So,  on  some 
trip  East,  they  could  stop  and  visit  company  x,  y,  and  z. 


Increasing  Commercialization  of  Academic  Biology 

Berg's  Prior  Refusal  of  Corporate  Consultantships 


Hughes:   You  at  Stanford  were  prime  scientific  movers  of  recombinant  DNA. 
You  must  have  been  aware  of  people  profiting  from  commercial 
application  of  recombinant  DNA  technology.   Was  there  motivation 
to  profit  from  it  yourselves? 


138 


Berg:     No,  not  at  all.   In  fact-- 


Berg:    --I  had  stayed  away  from  any  kind  of  consultantships,  even  though 
many  companies  had  come  to  me  and  asked  me  to  be  a  consultant. 
And,  I  think  they  had  probably  done  that  to  Arthur  as  well.   And 
except  for  Arthur's  connection  with  ALZA,  which  was  largely 
through  his  friendship  with  [Alejandro]  Zaffaroni  and  whatever 
other  little  companies  Zaffaroni  created,  Arthur  didn't  have  any 
consultantships,  as  far  as  1  know,  with  any  other  companies  other 
than  those  related  to  Zaffaroni. 

I  had  a  second  reason  for  not  accepting  consultant 
positions.   I  had  been  a  central  figure  in  the  "ethics  debate," 
the  safety  debate,  and,  given  the  public  policy  debate  that  was 
going  on  in  1975,  '76,  '77,  I  thought  I  would  lose  my  credibility 
in  terms  of  the  position  that  I  took  if  I  was  also  involved  in 
some  commercial  enterprise. 

It  seems  obvious  that  if  you're  working  for  a  company,  as  I 
see  now,  you  have  certain  obligations  or  interests,  and  those  can 
easily  influence  and  color  your  decisions  about  whether  you  think 
some  things  are  right  or  wrong,  appropriate  or  inappropriate,  and 
so  on.   So  I  decided  I  did  not  want  to  do  it.   I  was  asked  by  Dave 
Baltimore  to  join  one  or  another  commercial  groups,  but  I  chose 
not  to. 

But,  then  I  began  to  think  a  little  differently  about  it, 
although  I  didn't  do  anything  commercial  until  1980.   Dave 
Baltimore  said,  "Look,  this  field  of  genetic  engineering  is  going 
to  blossom  and  is  going  to  move  ahead.   If  you  participate  in  it, 
you  have  a  greater  chance  of  influencing  that  it  does  it  right  and 
properly  and  ethically  than  if  you  stay  out  of  it."   I  agreed  that 
was  a  reasonable  argument.   But,  nevertheless,  I  still  said  no  to 
most  things. 


Berg  Reconsiders  Corporate  Connections 

Berg:    It  was  probably  about  that  time  that  the  whole  DNAX  [Research 

Institute]  concept  got  started,  and  we  can  go  into  detail  about 
that.   But  that  was  the  first  time  that  I  at  least  conceded  to 
myself  that  this  was  an  interesting  proposition.   Probably  had  it 
not  been  Zaffaroni  [making  the  proposal  to  found  DNAX] ,  I  would 
have  said,  "No  way."  And  had  it  not  been  in  collaboration  with 


139 


[DNAX  co-founders]  Arthur  and  Charlie  Yanofsky,  I  would  have  said 
no. 

So  it  had  a  different  flavor.   Here  were  colleagues  1  really 
respected,  had  worked  well  with,  with  the  guy  who  was  going  to  do 
the  business  for  us,  who  was  a  man  I  admired  in  many  ways,  and  so 
I  finally  said  okay.   So  DNAX  was  the  first  time  I  conceded  that 
my  stand-offish  attitude  was  probably  outlived.   By  that  time,  the 
[biohazard]  debate  had  more  or  less  disappeared;  in  1980  things 
were  blossoming. 


The  Recombinant  DNA  Controversy 


Berg:     In  the  summer  of  [1976]  when  the  guidelines  were  issued,  it  was  a 
really  hot  debate.   I  was  traveling  around  the  country,  meeting 
with  city  councils  and  with  university  boards,  trying  to  put  a  lid 
on  preemptive  strikes  at  each  of  these  places  to  foreclose  the 
possibility  of  doing  recombinant  DNA  research,  in  the  city  of  San 
Diego,  for  example,  or  the  University  of  Michigan  campus.   I  was 
involved  in  all  these  forums.   So  during  that  period,  I  felt  it 
was  inappropriate  to  be  involved  in  corporate  ventures  and  didn't 
get  involved. 


Herbert  Boyer  and  Genentech 


Berg:    But  I  also  defended  the  guys  who  did,  particularly  Herb  Boyer.   I 
thought  there  was  an  opportunity  to  use  the  technology  to  do 
something  that  would  be  medically  significant,  and  if  Herb  wanted 
to  spend  his  time  to  do  that,  fine.   The  only  thing  I  criticized 
Herb  Boyer  on,  and  a  number  of  other  people  at  UCSF,  was  that  for 
a  brief  interval  after  they  founded  Genentech,  they  were  doing 
Genentech 's  work  at  UCSF  labs.   I  thought  that  was  totally 
inappropriate.   And  there  was  a  whole  lot  of  stuff  about  guys 
taking  their  materials  from  refrigerators.1  There  was  talk  of  a 


1  In  1999,  Genentech  and  UCSF  settled  a  case  out  of  court  regarding 
use  of  biological  material  allegedly  stemming  from  UCSF  in  Genentech 
recearch  on  human  growth  hormone. 


140 


kind  of  warfare  that  went  on  at  UCSF  amongst  Bill  Rutter,  Boyer, 
and  their  postdocs.1  That  I  thought  was  bad. 

Hughes:   Because  it  was  so  intimately  mixed  up  with  the  academic? 

Berg:    That's  right.   Because  they  were  doing  work  for  their  companies  in 
academic  centers.   That  was  totally  inappropriate.   But  once  the 
Genentech  labs  got  built,  and  they  had  a  place  to  work  off  campus 
and  so  on,  I  didn't  have  any  problem  with  that.   There  was  always 
the  possibility  of  a  fuzziness  in  the  boundary  between  what  was 
done  at  UCSF  and  what  was  being  done  at  Genentech  and  how  much  was 
passing  from  one  place  to  the  other.   And,  from  the  university's 
point  of  view,  they  were  losing  "intellectual  property"  by  people 
essentially  walking  away  with  it. 

Hughes:   How  did  you  feel  about  that  part  of  it? 

Berg:     I  spoke  out  against  it.   That's  the  only  area  on  which  I  was 
critical  about  the  people  who  were  moving  in  that  direction. 

Hughes:   Do  you  have  any  opinion  why  UCSF  happened  to  be  so  active  in 
technology  transfer? 

Berg:     Well,  Herb  Boyer  was  clearly  one  of  the  leaders  in  the  development 
of  the  technology  in  the  field  and  Bill  Rutter  was  one  of  the 
leading  people  in  biochemistry. 

Hughes:   So,  do  I  gather  from  that  comment  that  it's  more  personality,  more 
individuals,  rather  than  the  institutional  context? 

Berg:     Oh,  yes. 

Hughes:   From  what  you  have  just  told  me  today,  Stanford  might  be  seen  as  a 
better  locus  for  commercializing  recombinant  DNA  because 
interaction  had  been  going  on  between  industry  and  academia  for 
decades.   Whereas,  to  my  knowledge,  there  is  not  much  of  that  at 
UCSF  prior  to  the  recombinant  work.   So,  it  boils  down  to 
individuals? 

Berg:    Oh,  yes,  there's  no  question.   Herb  Boyer  being  approached  by 

[Robert]  Swanson  was  the  catalyst.   I  don't  know  what  went  through 
Herb's  head  in  regard  to  why  he  wanted  to  do  it.   Either  Swanson 
was  extremely  persuasive,  which  he  could  be,  or  Herb  wanted  to  get 
involved  with  something  to  make  money.  And,  I  think  Herb's 


1  For  coverage  of  this  controversy,  see:   Stephen  S.  Hall,  Invisible 
Frontiers:  The  Race  to  Synthesize  a  Human  Gene  (Redmond,  WA:  Tempus  Books, 
1987). 


childhood  and  background,  a  small  town  in  Pennsylvania,  is  often 
cited;  becoming  rich  and  famous  was  something  that  any  young 
American  kid  would  aspire  to.   I  don't  know  the  reasons.   But  he 
certainly  has  cited  that. 

This  is  just  an  aside,  but  if  you  look  at  what  Herb  has  done 
with  his  money,  he  has  done  some  really  tremendous  things  —  gifts 
to  UCSF,  to  Yale,  and  so  on.   But,  he  also  enjoys  collecting 
classic  cars  and  has  a  big  art  collection.   So  he  has  enjoyed  the 
wealth,  for  sure,  and  used  it  well  in  many  ways. 


William  J.  Rutter 


Berg:    Bill  Rutter  was  a  very  entrepreneurial  guy  right  from  the 

beginning.   1  knew  Bill  when  he  was  the  treasurer  of  the  American 
Society  of  Biochemistry  and  I  was  the  president  of  the  society. 
Bill  was  always  into  finance.   He  loved  it.   He  was  the  link 
between  the  society  and  the  people  who  were  investing  its  funds. 
He  was  keeping  track  of  our  budget  statements  and  how  much  money 
it  was  costing  us  to  run  the  journal.   Bill  was  an 
entrepreneurial,  financially  minded  guy.   He  understood  the  stock 
market.   And,  he  was  a  good  scientist.   There  is  no  question  that 
he  did  an  unbelievably  magnificent  job  of  resurrecting 
biochemistry  at  UCSF.   1  suspect  that  early  on,  he  also  saw 
opportunities  to  apply  what  he  knew. 


Cetus  Corporation 


Berg:     Cetus  was  another  company  that  pre-existed  recombinant  DNA.   It 

was  founded  by  Josh  Lederberg  and  by  Don  Glaser.   They  also  had  a 
very  entrepreneurial  businessman  who  probably  drove  that. 

Hughes:   Ronald  Cape. 

Berg:    Yes.   And  we  watched  that.   And  at  that  time,1  there  was  no 

recombinant  DNA.   What  they  were  trying  to  do  was  use  Glaser 's 
skill  at  instrumentation  to  create  a  machine  that  could  do  very 
rapid  screening  and  isolate  mutants  that  were  more  efficient  in 
producing  antibiotics  and  various  kinds  of  other  activities.   But 


1  Cetus  was  founded  in  1971,  before  the  discovery  of  recombinant  DNA 
cloning  technology. 


142 

they  had  nothing  to  do  with  recombinant  DNA  at  that  point.   But 
when  the  recorabinant  DNA  thing  came  on,  they  quickly  latched  onto 
it.   They  set  up  Stan  Cohen  here  with  Cetus  Stanford,  and  created 
something  called  Cetus  Immune  with  Hugh  McDevitt,  1  believe. 

Hughes:   Yes,  and  wasn't  there  an  agricultural  branch  of  Cetus? 
Berg:    In  Madison. 

They  set  up  Stan  here  somewhere  in  Palo  Alto,  I  think.  I 
don't  recall  where  Cetus  Immune  was  located.  As  near  as  I  can 
tell,  they  all  failed. 

Hughes:   You  don't  know  why? 
Berg:     I  don't  know  why. 

Hughes:   Was  Cetus  one  of  the  first  companies  founded  to  commercialize 
biological  knowledge? 

Berg:     Amgen  got  started  after  the  introduction  of  recombinant  DNA.   So  I 
think  Cetus  is  the  first.   Josh  was  still  here  as  chairman  of 
genetics.   I  don't  know  of  any  other  biology-based  company.   But 
there  may  be. 

Hughes:   Syntex? 

Berg:    Syntex  was  a  more  traditional  pharmaceutical  company  that  moved 
here  from  Mexico.   Zaffaroni  was  not  an  academic.   But  Carl 
Djerassi  had  worked  at  Syntex,  and  when  he  was  recruited  to  come 
to  Stanford  he  maintained  his  connection  with  Syntex. 

Hughes:   Is  Cetus  really  the  first  company  founded  by  biological 

scientists,  not  necessarily  by  people  applying  recombinant  DNA? 

Berg:     Except  for  Syntex,  I  don't  know  of  any  other. 

Hughes:   Was  it  significant  for  the  biotechnology  industry  that  a  company 
in  the  Bay  Area  was  making  a  go  of  applying  biology  in  the 
commercial  world? 

Berg:    I  don't  remember  that  the  creation  of  Cetus  and  its  operation  ever 
elicited  some  kind  of  admiration  or  emulation.   It  was  really 
making  use  of  Glaser's  skill  in  gadgetry,  in  invention  of 
machines,  because  all  they  were  doing  was  just  mechanizing  and 
automating  large-scale  ways  of  looking  at  microbial  colonies  and 
examining  them  in  some  clever  way. 


143 


I  remember  Don  Glaser  came  and  gave  a  seminar  and  got  a  big 
yawn.   He  was  a  very  bright  guy,  very  dynamic.   But  nobody  that  I 
recall  ever  said,  "Hey,  that's  the  wave  of  the  future." 


Stanford's  Policy  on  Consulting 


Policy  Reassessment,  1977 

Hughes:   Stanford  had  a  faculty  consulting  policy  which  well  predated  the 
recombinant  controversy. 

Berg:     Oh,  yes. 

Hughes:   But  in  March  of  1977,  President  Richard  W.  Lyman  delegated  to  the 
Board  of  Trustees  the  task  of  establishing  limits  for  consulting 
activities.1   I  can't  tell  you  whether  or  not  before  that  it  was 
university  policy  that  faculty  spend  not  more  than  one  day  a  week 
consulting.   But  certainly  that  came  to  be  by  1977.   Was  the  1977 
policy  prompted  by  what  was  happening  in  the  biological  sciences, 
or  was  this  a  more  general  problem  at  Stanford,  not  connected  with 
recombinant  DNA? 

Berg:    I  don't  remember  any  problems,  certainly  not  in  the  biological  or 
biomedical  area.   If  there  was  any  concern,  my  guess  is  that  Lyman 
would  have  been  more  concerned  about  the  engineering  school,  which 
had  traditionally  done  a  lot  of  consulting.   There  were  also 
people,  for  example,  in  the  education  field  who  were  doing  a  lot 
of  consulting  in  various  areas.   1  don't  remember  that  there  was 
any  specific  event  that  instigated  Lyman  to  do  that. 

Hughes:   I'm  glancing  through  the  document  and  I  don't  see  anything  about 
why  Lyman 's  concerned. 

Berg:    He  says  the  policy  contains  some  ambiguities.   "It  is  in  the  best 
interest  of  both  the  faculty  and  the  University  to  clarify  these 
ambiguities. . ."2 

Hughes:   There  must  be  something  provoking  him  to  reassesss  the  policy. 


1  Lyman  to  members  of  the  Academic  Council,  March  18,  1977  (Arthur 
Kornberg  papers,  SC  359,  Green  Library,  Stanford,  box  5,  folder:  1977). 

2  Ibid. 


144 

Berg:    Well,  I  think  what  happened,  and  maybe  it  happened  around  that 

time.   There  was  a  guy  at  UC  Davis- 
Hughes:   Ray  Valentine? 

Berg:     Valentine,  who  helped  set  up  a  plant  biotech  company. 
Hughes:   Calgene. 

Berg:    Yes.   There  was  a  bit  of  a  furor  over  the  fact  that  Calgene  gave 
Valentine  a  research  grant.   He  was  one  of  the  founders  and  the 
principle  officer  in  the  company,  and  they  gave  him  a  grant  to 
carry  on  the  research  on  the  campus,  that  is,  in  a  university  lab. 
That  elicited  a  certain  amount  of  eyebrow  raising—was  that 
appropriate?   My  recollection  is  that  there  was  a  certain  amount 
of  clucking  that  went  on  about  that. 

I  think  the  University  of  California  established  a  policy 
which  was  that  faculty  could  not  accept  grants  from  companies  in 
which  they  had  a  financial  interest.   And  it  may  be  that  was  one 
of  the  things  that  stimulated  Lyman.   Maybe  that  was  one  of  the 
ambiguities.   I  don't  have  the  chronologies--  When  did  Cetus 
Immune,  and  when  did  Cetus  Palo  Alto  get  created?  The  question 
was,  was  this  a  way  of  funding  Stan  Cohen's  research,  or  Hugh 
McDevitt's?   I  think  there  may  have  been  a  little  bit  of  a  concern 
about  this  boundary  between  your  own  lab  and  the  lab  that  you  are 
presumably  directing  for  a  company. 

Hughes:   Well,  certainly,  Donald  Kennedy  was  very  concerned,  and  even 

testified  in  the  House  in  1981.   He  says  in  his  testimony  that 
Stanford's  faculty  Committee  on  Research  had  voted  overwhelmingly 
to  reject  the  university's  equity  participation  in  faculty 
research  ventures.1 

Berg:    I  was  one  of  the  people  that  met  with  a  group  of  venture  capital 
people,  university  people,  and  trustees  to  discuss  this  issue  of 
whether  the  university  should  in  fact  get  into  the  business  of 
venture  capital.   Should  it  be  using  its  funds  to  help  faculty 
found  businesses?   I  argued  vehemently  against  it.   I  wasn't  the 
only  one.   The  venture  capital  people  urged  the  university  not  to 
do  it,  saying  that  the  university  didn't  understand  it;  it  would 
be  a  disaster  for  them. 


1  Statement  of  Dr.  Donald  Kennedy,  President,  Stanford  University, 
Stanford,  California.   Subcommitee  on  Investigation  and  Oversight, 
Committee  on  Science  and  Technology,  House  of  Representatives, 
Commercialization  of  Academic  Research,  June  8  &  9,  1981,   U.S.  Government 
Printing  Office,  pp.  6-28. 


145 


But,  more  important,  we  were  more  worried  about  the  impact 
on  the  academic  setting.   When  the  university  is  in  business  with 
one  of  its  faculty,  what  kinds  of  relationships  does  that  create? 
Does  a  guy  get  more  money?   Does  a  guy  get  favored  promotions? 
There  were  all  kinds  of  things. 

But,  going  back  to  '77,  again  without  knowing  the  chronology 
of  when  these  companies  were  popping  up  around  here.   I  don't  know 
of  anybody  else  in  1977  who  was  doing  any  consulting.   What  did 
get  prominence  was  in  fact  that  here  we  had  on  our  campus  or  in 
our  vicinity  two  of  our  professors  engaged  in  a  company.   Cetus 
was  already  an  existing  company.   If  somebody  felt  there  was  going 
to  be  some  financial  gain,  the  way  to  do  it  was  to  start  a 
spinoff,  start  something  from  scratch,  so  a  person  got  in  on  the 
ground  floor  for  any  financial  rise.   So,  I  think  that  was  one 
motivation  for  creating  those  two  companies,  or  the  three. 

The  third  was  to  place  new  companies  in  the  locale  where 
those  people  were  doing  their  research,  because  they  could  easily 
have  said,  "Look,  1  haven't  got  time  to  go  over  to  the  East  Bay 
and  solve  this  problem,"  and  so  on  and  so  forth.   "But,  if  it's 
right  in  my  backyard,  I  would  be  left  to  run  the  operation."   1 
suspect  that  is  some  of  their  motivation. 


The  Shooter  Committee  on  Conflicts  of  Interest 


Hughes:   I  saw  a  reference  to  the  Shooter  Committee. 

Berg:     I  was  on  the  Shooter  Committee.   The  Shooter  Committee  was  one  set 
up  by  the  medical  school. 

Hughes:   It's  called  the  Shooter  Committee  on  Conflicts  of  Interests.   So, 
you  see  there  really  is  a  lot  of  turmoil  around  this  topic. 

Berg:    That's  right.   You  have  to  recognize  that  the  previous  academic 
environment  was  very  permissive.   Nobody  kept  track  of  anything. 
Nobody  knew  what  affiliations  anybody  had  with  companies.   Nobody 
checked  on  how  much  time  they  spent  doing  these  things.   So,  it 
was  really  an  attempt  to  step  back  and  say  look,  something  is 
happening;  our  faculty  is  becoming  involved  [commercially],  and 
have  been  involved,  i.e.  the  engineering  people  and  chemistry 
people. 

Maybe  the  fact  that  the  University  of  California  put  a  lid 
on  one  area  of  involvement  energized  Stanford  to  look  at  the  whole 
problem.   And  so  Lyman  reiterated  what  he  felt  was  the  policy.   It 


146 

wasn't  that  stringent.   It  said  that  you  could  consult  for  20 
percent  of  your  time. 

Hughes:   One  of  the  tensions  was  that  the  clinical  people  had  to  turn  over 
their  consulting  fees  to  the  medical  school.   There  was  no  control 
of  the  nonclinical  faculty,  as  you're  saying;  they  just  put  the 
consultant  fee  in  their  pocket;  that  was  it. 

Berg:    That's  right.  Absolutely.   Now,  you're  ringing  a  bell.   Henry 

Kaplan,  who  was  head  of  radiology  and  was  one  of  the  most  powerful 
and  influential  figures  in  the  medical  school,  was  always  griping 
about  the  fact  that  clinical  faculty  could  not  accept  fees  or 
anything  of  that  sort.   They  were  expected  to  be  full-time,  and  as 
full-time  faculty,  they  were  expected  to  turn  over  all  earnings  to 
the  medical  school.   And,  I  remember,  as  chairman  of  the 
department,  almost  every  executive  committee  meeting  was  Kaplan 
pounding  on  that  inequity.   He  kept  identifying  how  inequitable  it 
was  that  basic  scientists  were  beginning  to  develop  these  kinds  of 
consulting  relationships  and  could  keep  their  consultant  fees. 

I  remember  Kornberg  made  this  argument,  "We'll  take  your 
salaries,  and  we'll  give  up  our  consulting  fees.   But  if  you 
consider  the  disparity  in  our  salaries,  you  can't  be  complaining 
about  that  we're  engaged  in  this  kind  of  consulting  activity." 
So,  that  sort  of  kept  a  lid  on  it. 

The  Shooter  Committee  had  to  really  look  at  the  whole  issue. 
I  was  a  participant  in  that  committee.   What  was  a  conflict  of 
interest?  A  conflict  of  interest  in  my  view  arose  if  you  have  a 
financial  stake  in  some  decision  you're  making,  and  you  haven't 
disclosed  that  you  have  a  financial  stake. 

But  the  more  critical  thing  from  our  point  of  view  was  what 
I  called  conflict  of  obligation.   In  other  words,  you're  a  faculty 
member  here;  you're  expected  to  be  here  full  time;  you're  being 
paid  a  salary.   You're  here,  and  we  expect  you  to  provide  the  kind 
of  intellectual  heat  and  creativity  that  makes  this  [university]  a 
great  place.   If  your  head  is  somewhere  else,  there  is  a  conflict 
of  obligation.   If  you're  thinking  more  about  something  you're 
doing  on  the  outside  than  what  you  are  doing  here,  then  you're 
lost  to  us  as  a  valuable  person. 

And  so,  we  had  to  look  at  the  whole  issue  of  what  was 
conflict  of  interest,  and  that  was  easy.   You  could  easily  say  you 
have  to  declare  any  financial  interest  with  anyone.   If  you're  in 
a  position  to  order  a  piece  of  equipment,  and  you  buy  it  from  a 
company  in  which  you  have  a  big  financial  stake,  that's  a  conflict 
of  interest.   But  conflict  of  obligation  was  much  more  subtle. 
For  example,  you're  traveling  around  the  country  all  the  time 


147 


giving  lectures,  you're  teaching  courses  at  university  X,  or  some 
summer  program,  and  you're  not  around  here  and  you  don't  fulfill 
your  teaching  responsibility  here.   That's  academic;  that's  not  in 
any  way  a  consultantship;  that's  not  in  this  20  percent  time. 

You  serve  on  many  boards ;  you  serve  on  many  government 
committees,  but  you're  not  around  campus.   So  the  question  is,  how 
do  we  view  that?  How  do  we  view  a  person  who  disappears  from  the 
scene  to  write  a  book?  Obviously,  he  gets  financial  rewards  from 
that  book.   What's  different  about  that  kind  of  activity  than 
somebody  who  goes  off  and  consults  for  a  company,  gets  paid  by  a 
company? 

Then,  the  second  thing  was,  should  we  be  snooping  into  how 
much  money  people  are  getting  for  their  consulting?  The  Shooter 
Committee  said  that  wasn't  our  business.   But,  we  put  in  place  a 
reporting  requirement,  that  is,  every  faculty  member  was  required 
to  report  what  their  outside  activities  involved  and  estimate  how 
much  time  they  spent  at  them.   And,  we  believed  that  by  forcing 
people  to  write  down  something,  they  would  be  lying  if  they  did 
more.   I  don't  think  anybody  wanted  to  lie  or  disobey,  but  when 
nobody  asked,  it  was  easy  to  just  say,  "Okay,  it's  a  little  bit 
more  this  month  than  last  month."  But  very  quickly,  you'd  find 
that  somebody  was  spending  a  lot  of  time  away.   But  we  also  set 
down  very  clearly  this  whole  concept  of  conflict  of  obligation.   I 
think  that's  now  part  of  the  lore  that's  used  in  terms  of 
evaluating. 

Hughes:   Well,  how  did  you  settle  that  conflict  of  obligation? 

Berg:    We  just  pointed  out  that  this  was  an  area  in  which  we  expected 

individuals  to  fulfill  their  obligation  to  the  university.   They 
were  being  paid  full  time  to  spend  their  time  here  and  contribute 
to  the  life  and  activities  of  the  university. 

I've  had  this  conflict  argument  with  other  people;  David 
Baltimore  and  I  have  argued  this.   He  probably  spends  30-40 
percent  at  MIT  and  60-70  percent  of  his  time  elsewhere  doing  great 
things.   He  said,  "I  am  carrying  the  banner  for  MIT.   By  my  being 
on  various  committees  and  so  on  and  so  forth,  I  am  representing 
MIT  and  fulfilling  an  obligation  to  keep  MIT  at  the  forefront." 
So,  people  had  different  perspectives  on  this. 

** 

Berg:    Some  believe  that  benefits  accrue  to  the  university  from  their 

participation  and  consulting,  bringing  the  outside  world  into  the 
ivory  tower,  knowing  where  the  cutting  edge  is,  what  some  of  the 
problems  are,  making  opportunities  available  to  their  students. 


148 


Things  like  that.   I  mean,  this  20  percent  time  has  been  justified 
in  many  different  ways,  and  it  exists  at  Harvard,  Yale,  and  MIT, 
and  most  other  [universities].   It  was  understood  that  consulting 
was  not  to  exceed  a  day  a  week,  but  nobody  kept  track,  nobody 
asked.   I  think  most  universities  now  require  faculty  to  fill  out 
an  accounting  of  whom  they  consult  for  and  roughly  how  much  time 
they  spend  at  it  each  year. 

Hughes:   And  who  sees  those  reports? 

Berg:    It  goes  to  the  dean.   But,  if  you  ask  me  does  the  dean  ever  look 
at  it;  does  somebody  in  the  dean's  office  look  at  it?   I  don't 
know.   They  keep  it  on  record.   I  think  it's  there  largely  should 
some  abuse  arise.   Then  someone  can  go  back  and  say,  "Look,  you 
said  that  this  year  you  were  not  going  to  spend  more  time 
[consulting].   Here,  we  now  find  out  that  you're  involved  in  about 
five  things.   You're  the  owner  of  a  company,  or  you're  co-founder 
of  a  company,  and  you're  spending  a  lot  of  time  as  a  company 
officer."   So,  it  was  largely  there  just  to  have  a  record,  which 
probably  could  be  used  when  abused.   But  I  don't  think  anybody 
actually  reviews  these  reports. 


IV   DNAX  RESEARCH  INSTITUTE  OF  MOLECULAR  AND  CELLULAR  BIOLOGY, 
INC. 


Earlier  Commercial  Ventures 

Hughes:   In  the  second  oral  history  that  you  did  for  MIT,  which  was  with 

Charlie  Weiner,  you  said- 
Berg:     What  year  was  that? 

Hughes:   1978.   You  did  an  earlier  one  in  1975. 
Berg:     Yes. 

Hughes:   --that  you  didn't  at  that  time  want  to  be  involved  with  a  company. 
I  quote,  "I  have  told  at  least  four  or  five  different  companies 
that  I  will  not  participate  as  a  consultant."1  DNAX  was  founded 
in  1980,  right? 

Berg:     Yes. 

Hughes:   How  did  you  get  involved? 

Berg:     Well,  I've  read  Arthur's  recounting  of  the  story.2   Both  Charlie 
Yanofsky,  and  I  have  a  slightly  different  story.   [laughter]   And 
it  could  be  because  we  weren't  aware  of  some  of  things  that  Arthur 
was  doing  or  saying.   First  of  all,  Arthur  was  a  consultant;  he 
was  actually  a  member  of  the  board  for  ALZA.   He  was  also  a  member 
of  the  board  for  some  little  spin-off  that  ALZA  created  to  try  to 
immobilize  food  dyes  on  various  things,  and  that  failed. 


1  Paul  Berg  (second  interview),  April  17,  1978,  p.  64.   MIT  Institute 
Archives,  Recombinant  DNA  Oral  History  Collection. 

2  Arthur  Kornberg.   The  Golden  Helix:  Inside  Biotech  Ventures. 
Sausalito,  California:  University  Science  Books,  1995. 


150 


Channing  Robertson's  Company 

Berg:    There  was  a  guy  named  Channing  Robertson  who  was  the  head  of  the 

chemical  engineering  department.   He  had  the  idea  that  the  biotech 
industry  was  going  to  eventually  want  to  produce  products. 
Everybody  was  cloning  things,  and  they  were  trying  to  figure  out 
how  to  get  them  expressed.   But,  in  the  end,  what  had  to  be 
produced,  if  it  was  going  to  be  usable,  had  to  be  produced  in 
large  quantities.  And,  being  a  chemical  engineer,  he  was  very 
tuned  into  how  you  go  about  manufacturing  chemicals . 

What  Robertson  realized  was  that  this  was  a  whole  new 
ballgame  because  proteins  were  going  to  be  the  products  of  these 
genes,  and  the  chemical  industry  had  not  had  any  experience  of  how 
to  produce  large  quantities  of  proteins.   So  he  had  been 
developing  the  kind  of  technology  which  allowed  one  to  be  able  to 
grow  bacteria  to  very,  very  high  densities,  much  higher  than  any 
way  you  can  grow  them  in  a  flask. 

He  could  see  that  technology  was  going  to  be  advantageous 
since  bacteria  at  that  time  semed  to  be  the  organisms  that  were 
going  to  be  producing  these  rare  proteins.   If  you  could  grow  them 
to  higher  and  higher  quantities--  He,  not  being  a  biologist, 
needed  some  help,  and  he  came  to  see  Kornberg.   He  also  talked  to 
Charlie  Yanofsky  who  was  very  experienced  in  growing  bacteria. 

Robertson  told  us  about  this  notion  he  had  of  forming  a 
company,  and  that  Stanford  was  in  some  way  interested  in  becoming 
a  partner.   Not  bankrolling  the  whole  thing,  but  in  fact  retaining 
some  kind  of  interest  should  this  company  do  well.   He  went  out  to 
a  lot  of  chemical  companies  and  raised  a  whole  lot  of  money.   I 
don't  think  Stanford  put  any  money  into  it,  but  Stanford  retained 
some  form  of  interest  which  I  just  can't  remember.   He  had  a 
venture  capital  firm  here  on  Sand  Hill  Road,  Palo  Alto  that  would 
be  a  principal  investor.   Robertson  came  to  us  and  asked  if  we 
would  co-found  this  company  with  him. 

Hughes:   We  being? 

Berg:    Kornberg,  me,  and  Yanofsky.  We  were  to  be  the  scientific  backing 
for  a  venture  into  the  next  stage  of  genetic  engineering.   Instead 
of  doing  it  in  test  tubes,  we  were  going  to  do  it  in  factories. 
And  we  said  okay.   This  was  about  the  time  when  I  began  to 
reconsider  my  earlier  objections  to  being  involved  in  commercial 
biotechnology.   Zaffaroni  was  not  even  involved  in  this  venture. 

Hughes:   Was  this  in  1980? 


151 


Berg:    Probably  the  year  before.   Could  have  been  '80,  but  I  think  '79 

more  likely.   We  thought  it  was  kind  of  an  interesting  opportunity 
and  that  Robertson  had  a  fascinating  idea  for  the  kind  of 
technology  he  wanted  to  develop.   He  was  the  engineering  expert. 
And  so  we  said  okay.   Then,  we  went  to  meet  with  the  venture 
capitalist  to  discuss  what  our  stake  would  be  in  this  company. 

We  were  so  sickened  by  our  discussion  with  this  guy  that  we 
came  out  of  there  just  saying,  "Pretty  slimy  character."  Although 
he  isn't  a  slimy  character,  I've  learned  since.   But  what  they 
were  willing  to  give  us  in  terms  of  stock  options  and  blah,  blah, 
blah,  seemed  marginal.   But,  since  we  had  never  been  involved  in 
this  kind  of  business  that  wasn't  so  clear. 


Alejandro  Zaffaroni 


Berg:    So  Arthur  went  to  ask  Alex  Zaffaroni  whether  that  was  a  fair  deal. 
That's  my  recollection.  Arthur  says  he  came  to  it  in  a  very 
different  way.   But  I  remember  that  Charlie  and  I  thought  of 
asking  Zaffaroni  whether  this  was  a  conventional  standard,  fair 
kind  of  grant.   And  Zaffaroni  just  laughed  and  thought  it  was 
ridiculous  and  said,  "If  you  guys  are  really  serious  about  wanting 
to  get  involved  with  a  biotech  company,  I'll  form  one  and  you  can 
join  me." 

Alex  then  told  Arthur  and  us  that  he  had  been  approached  and 
had  actually  considered  becoming  an  investor  and  a  guiding  light 
for  a  biotech  company  that  Harvard  might  start.   He  had  been 
approached  and  asked  whether  he  would  be  interested.   He  had 
consulted  with  Arthur  about  it—which  may  be,  but  certainly  we 
were  unaware  of  that  conversation- -but  he  said  he  turned  them 
down.   When  he  realized  that  we  were  interested  he  began  to 
consider  doing  something  with  us. 

I  didn't  really  know  Zaffaroni  well,  but  I  knew  him  through 
Arthur,  and  I  knew  Arthur  had  the  highest  regard  for  him.   He  was 
clearly  a  very  innovative  and  creative  entrepreneurial  guy.   He 
said  he  would  do  all  the  business,  and  what  he  wanted  us  to  do  was 
to  provide  the  scientific  energy  behind  forming  this  company. 

Zaffaroni  may  have  told  us  what  our  financial  stake  would  be 
if  we  started  this  company.   It  put  the  other  one  to  shame.   The 
same  attraction  that  had  led  the  three  of  us  to  even  consider 
joining  with  Channing  Robertson  was  there  in  thinking  about  DNAX, 
except  that  now,  with  Zaffaroni,  we  had  somebody  whom  we  respected 
and  had  confidence  in.   The  Robertson  company  seemed  more 


152 

ephemeral,  although  there  was  going  to  be  a  lot  of  other  companies 
involved  along  with  the  university.   I  don't  remember  that  I 
particularly  thought  that  was  bad  because  their  stake  was  not 
anything  like  what  I  had  argued  against  before.   It  was  a 
different  nature.  Again,  I  don't  remember  the  details. 

Hughes:   So  you  got  out  of  the  arrangement  with  Robertson? 
Berg:    We  got  out  of  it. 
Hughes:   Did  anything  happen? 

Berg:    We  went  to  Channing  and  told  him  that  we  had  this  alternative 

opportunity,  and  we  were  much  more  attracted  to  that.   For  one,  it 
got  us  into  an  area  that  was  more  closely  related  to  our 
interests,  which  was  how  to  use  recombinant  DNA  to  make  a 
significant  medical  product,  rather  than  commercial  quantities  of 
proteins.   Robertson's  company  was  formed  and  went  along  for  a 
little  while,  and  then  it  failed.   I  think  the  companies  that  were 
interested  from  the  beginning  began  to  recognize  that  there  were 
other  kinds  of  technology  available  to  achieve  the  same  ends, 
whatever.   But  it  eventually  failed. 


DNAX 


Initial  Research  Focus,  Recruitment,  Science  Advisors 


Hughes:   Had  DNAX  been  refined  any  further  than  that?   What  were  you  hoping 
to  do  with  recombinant  DNA  technology? 

Berg:    Well,  we  had  several  brainstorming  sessions.   And  we  decided  that 
immunology  would  be  the  focus.   Once  we  were  engaged,  Alex 
Zaffaroni  said,  "I  want  to  bring  in  a  bunch  of  other  people,"  one 
of  whom  was  a  guy  named  Ed  Haber,  who  just  recently  died.   He  was 
a  professor  at  Harvard.   And  then  he  engaged  a  bunch  of  people  who 
had  been  involved  in  ALZA,  about  five  or  six  other  people. 

The  first  question  was,  who  were  going  to  be  the  scientists? 
We  were  implored  to  try  to  convince  some  of  our  postdocs  to  join 
DNAX.   The  first  group  we  approached  were  postdocs,  the  first 
being  Kenichi  Arai,  from  Arthur's  lab.   He  had  probably  been 
Arthur's  best  postdoc;  he  was  a  dynamo.   But  he  had  gone  back  to  a 
position  in  Japan  but  his  wife,  who  was  also  a  scientist,  couldn't 
have  a  position  there.  Arthur  contacted  him  and  said,  "How  would 
you  like  to  come  back?"  He  did.   There  was  a  postdoc  of  Charlie 


153 


Yanof sky's,  Gerard  Zurowski,  who  had  gone  to  Australia  with  a 
Queen's  Fellowship.   He  came  back.   Lee  Hood,  I  think,  was  an 
advisor.   So  he  sent  us  one  of  his  people,  Kevin  Moore,  who  is 
still  at  DNAX. 

Hughes:   It  was  an  impressive  group  of  advisors. 

Berg:    This  is  typical  of  Alex  Zaffaroni;  you  engage  a  lot  of  big  names, 
to  impress  potential  investors  and  for  recruiters.   But  a  lot  of 
the  advisors  contributed  very  little. 


Ed  Haber  and  the  Engineering  of  Monoclonal  Antibodies 


Berg:    The  interesting  thing  was  that  Ed  Haber  was  a  cardiologist  at 

Harvard.   One  of  the  drugs  that  he  pointed  out  was  very  useful, 
was  Digoxin.   Older  people,  he  said,  frequently  overdose  on 
digoxin.   They  forget  that  they  just  took  their  pill  and  they  take 
another.   Or,  some  kids  eat  grandpa's  pills  and  they  go  into  coma. 
What  he  had  done  in  order  to  treat  people  like  this  was  to  develop 
a  monoclonal  antibody,  which  he  was  using  to  inject  into  such 
people  with  remarkable  results;  like  within  twenty  minutes  to  a 
half  an  hour,  they  would  be  out  of  the  comatose  state,  get  off  the 
bed,  and  go  home. 

Hughes:   Where  would  he  have  found  the  monoclonal  antibody? 

Berg:     He  made  it.   He  was  an  immunologist  by  training,  a  very  good  one. 
But  he  was  a  cardiologist  by  profession.   So  he  made  a  monoclonal 
antibody  against  Digoxin  by  attaching  Digoxin  to  an  inert  carrier. 
You  inject  the  carrier  and  then  you  screen  for  antibodies  that  are 
directed  against  the  chemical  entity  to  which  you  attach  it.   You 
remove  all  the  antibodies  that  are  to  the  inert  carrier,  and  then 
you  get  antibodies  very  specific  against  Digoxin.  And  then,  you 
try  to  get  one  that  has  a  very  high  affinity  for  it. 

What  Haber  found  was  that  he  could  clear  the  body  of  Digoxin 
by  just  injecting  the  monoclonal  antibody.  Antibodies  come  with 
two  kinds  of  chains,  heavy  and  light  chains.   He  claimed  that  you 
could  take  the  Digoxin  antibody  apart  and  it  would  reassemble.   I 
think  he  also  had  shown  that  you  could  make  what  they  call  an  Fab 
fragment,  which  is  a  fragment  of  the  antibody  but  it  contains  the 
combining  sites.   Fab  fragments  have  all  the  same  kind  of 
specificity  and  affinity  for  these  ligands,  antigens.   But  they're 
much  smaller  and  they  are  secreted  or  excreted  very  rapidly. 


154 

Haber  had  shown  that  the  Fab  fragments  were  even  more 
efficient  in  being  able  to  sop  up  the  digoxin.   They  gained  entry 
into  places  that  the  big  proteins  can't  go.   They're  smaller  bits; 
they  go  in  and  attach  and  eventually  are  excreted.   So,  we  thought 
we  could  synthesize  the  heavy  and  light  chains  in  bacteria  by 
isolating  the  genes  that  encode  the  heavy  and  light  chain 
proteins.  And,  since  he  had  these  cells  that  were  making  this 
monoclonal  antibody,  we  could  isolate  the  cDNAs  from  them  and 
engineer  them  so  that  we  could  make  heavy  and  light  chains  in  £_._ 
coli.   And,  the  heavy  and  light  chains  could  be  prepared  in  large 
quantities,  then  we  could  reconstitute  the  active  antibody. 

We  knew  it  wasn't  possible  to  engineer  the  structure  of  the 
antibody  combining  site  directly,  but  we  could  change  the 
structure  of  the  gene  at  will.   And  we  could  make  antibodies  with 
very  different  kinds  of  specific  affinities.   So,  the  concept  had 
emerged- -Zaffaroni  gave  it  a  name,  but  I  can't  remember  the  name 
of  it.   But  you  could  imagine  that  you  could  start  making 
antibodies  that  would  be  used  in  the  chemical  industry  for 
extracting  things.   You  could  make  antibodies  specific  to 
anything.   You  could  use  them  like  a  first-aid  kit.   You  can 
imagine,  for  every  kind  of  toxic  substance  that  somebody  could 
ingest  there  could  be  a  monoclonal  antibody  that  would  be  injected 
into  a  person  to  neutralize  the  toxic  agent. 

Hughes:   How  far  along  was  the  technology?  How  grounded  in  actual  science 
were  all  these  ideas? 

Berg:    We  knew  you  could  make  cDNAs .   We  had  these  brainstonning 

sessions.   We  sat  around  the  table  and  said,  "Okay,  our  first 
target  ought  to  be  to  use  genetic  engineering  to  make  specific 
antibodies,  or  parts  of  antibodies,  engineered  in  ways  to  give 
them  special  properties,  which  could  be  used  in  a  variety  of 
commercial  and  industrial  and  medical  uses." 


Utilizing  the  Okay ama -Berg  Procedure 


Berg:    Now,  my  lab  had  just  worked  out  a  technique  for  being  able  to  make 
full-length  cDNAs.   The  earlier  work  that  had  been  done  by  Tom 
Maniatis  said  made  it  possible  to  make  cDNAs  but  they  were  rarely 
full-length,  full-length  meaning  that  they  contained  the  entire 
protein  coding  sequence. 

Hughes:   Would  that  be  Okayama? 


155 

Berg:    Yes.   The  Okayama-Berg  procedure  was  designed  to  guarantee  that 

you  got  full-length  cDNAs,  even  if  they  were  very  big  genes.   And 
so  I  said,  "We  have  a  technique  for  being  able  to  use  Haber's 
cells,  extract  the  RNA  from  them  and  make  full-length  cDNAs  that 
would  encode  the  heavy  and  light  chains."  Charlie  Yanofsky  was 
the  world's  expert  on  regulating  expression  of  genes  in  E.  coli. 
We  could  then  engineer  these  coding  sequences  into  E.  coli  and 
make  large  quantities  of  heavy  and  light  chains.   Then,  we  would 
extract  these  heavy  and  light  chains,  purify  them,  then  put  them 
together,  and  then  reform  the  antibody. 

Hughes:   Was  it  somewhat  fortuitous  that  you  three  had  major  facets  of  the 
problem  to  contribute?   Is  that  why  you  were  involved  initially  in 
DNAX? 

Berg:    When  we  agreed  with  Zaffaroni,  there  was  no  clear  plan  of  what  we 
were  going  to  do.   Therefore,  our  individual  expertise,  other  than 
being  in  a  general  area  of  molecular  biology  and  molecular 
genetics,  was  not  evident.   It  was  Ed  Haber  who  brought  this  as  a 
possibility  and  said  there  was  a  market  for  these  kinds  of  things. 
Then  we  began  to  say,  well,  how  would  we  make  antibodies?   Well, 
the  only  way  to  make  antibodies  that  I  knew  was  to  actually  clone 
the  genes.   But  not  the  genes  because  the  genes  are  much  more 
complicated  to  work  with.   Clone  the  cDNAs,  express  them  in  £_._ 
coli ;  Charlie  knew  how  to  do  that.  Arthur  was  sort  of  not 
involved  because  he  wasn't  an  expert  in  any  of  these  areas.   But 
we  did  adopt  the  Okayama-Berg  method  as  the  principle  approach  to 
do  this. 

Okayama  being  Japanese  sometimes  writes  English  a  little 
more  like  Japanese.   And  so,  the  method  that  was  published  was 
actually  quite  difficult  for  most  people  to  follow  because  it  was 
quite  intricate.   Again,  like  the  first  creation  of  the  first 
recombinant,  it  required  a  lot  of  experience  with  enzymes  and 
dealing  with  them  in  different  ways.  Most  of  the  enzymes  we  had 
in-house . 

Hughes:   So,  you  had  a  corner  on  the  market? 
Berg:    That's  right. 


Problems  in  Engineering  Monoclonals 


Berg:    Kenichi  Arai  came  from  Japan.   He  immediately  consulted  with 

Okayama,  and  before  you  knew  it,  these  guys  at  DNAX  were  cranking 
out  clones  with  no  problem.   It  turned  out,  they  made  heavy  and 


156 


light  chains,  but  they  would  not  reassociate.   Ed  Haber  had  shown 
that  he  could  reassociate  heavy  and  light  chains  for  other 
antibodies,  but  not  this  one.   So  here  we'd  gone  through  this 
entire  exercise  and  everything  worked  perfectly;  full-length  cDNAs 
were  being  expressed  in  E.  coli.   But,  they  would  not  reassociate. 
We  tried  to  make  it  so  that  both  genes  were  being  expressed  in  the 
same  cell,  and  maybe  they  would  reassociate  under  those 
conditions.  We  never  could  make  a  functional  antibody. 

Money  was  running  out.  Alex  had  raised  some  $4.5  million  to 
get  DNAX  started.  We  didn't  have  more  than  about  ten  or  twelve 
scientists  working  and  we  had  used  up  all  of  this  money. 


Fundraising 

Hughes:   I  know  Arthur  was  involved  in  some  fundraising  trips. 

Berg:     Oh,  yes. 

Hughes:   Were  you  as  well? 

Berg:    I  went  on  one  fundraising  trip  with  Alex  to  New  York,  where  we  met 
with  the  people  from  the  Rothchild  venture  capital  company.   It 
was  based  in  New  Jersey.   Alex  and  I  met  with  their  group  of 
scientific  advisors;  tried  to  get  them  to  invest.   Sydney  Brenner 
was  their  consultant.   He  put  us  at  the  top  of  the  heap  in  terms 
of  scientific  expertise  and  prominence.   He  thought  DNAX  was  a 
great,  great  idea,  and  so  on  and  so  forth.   But,  they  never  bit. 
In  fact,  it  was  one  of  the  greatest  disappointments  I  ever  had. 
We  never  even  got  a  response  from  them.   We  met  with  them,  we  took 
them  to  dinner,  and  we  had  this  fancy  talk.   I  thought  we 
convinced  them  and  that  they  were  going  to  buy  it.   [laughter] 
But,  we  never  even  got  a  reply. 

Hughes:   Do  you  think  they  understood  the  science? 

Berg:    Oh,  yes,  they  understood  the  science.  Arthur,  I  think,  met  with 
somebody  from  Rothchild 's  on  one  of  his  trips  to  England.   We 
never  could  find  out  why  it  didn't  go.   But,  in  any  case,  Arthur 
and  Alex  made  several  trips  to  Japan  where  Arthur  had  all  kinds  of 
contacts.  A  former  postdoc  of  his,  Omura,  was  a  major  figure  for 
Takeda  Chemical.   So  DNAX  was  on  the  verge  of  going  under. 


157 
Schering-Plough  and  DNAX 

Schering-Plough 's  Research  History 

Berg:    I  think  Alex  went  to  Schering-Plough  because  the  new  management  of 
Schering-Plough  were  people  that  he  had  known  from  Ciba-Geigy. 
Ciba-Geigy  had  been,  essentially,  the  owner  of  ALZA;  they  bought 
ALZA  from  him.   And  so  he  went  to  see  them  to  tell  them  about 
DNAX.   And  there  was  the  almost  extraordinarily  fortuitous 
circumstance  that  they  were  looking  for  new  directions.   The 
company  had  been  at  its  nadir  in  terms  of  successful  products. 
They'd  had  a  very  successful  antibiotic,  which  was  going  off 
patent.   Their  prospects  were  zilch;  they  had  nothing  in  the 
pipeline.   And,  they  had,  during  the  year  1978- '79,  conducted  a 
strategic  survey  of  what  were  the  promising  directions  to  go  in  in 
the  future.   And,  they  chose  immunology. 

To  implement  this  new  strategy  in  immunology,  they  created  a 
laboratory  in  Lyons,  France,  largely  because  they  were  obliged  to 
by  the  French  government  in  order  to  protect  their  pricing 
capabilities.   So  they  built  a  lab  there  and  said  this  is  where 
we're  going  to  do  immunology.   And  when  they  went  out  to  try  and 
recruit  people  to  do  it,  they  were  really  unsuccessful. 
Everything  that  they  tried  to  bring  immunologists  into  their 
research  organization  had  failed,  in  part  because  their  reputation 
was  pretty  lousy  for  research. 

So  when  Zaffaroni  presented  them  with  the  existence  of  a 
company  committed  to  immunology,  and  they  saw  the  board,  and  they 
saw  the  scientific  advisors,  and  they  saw  that  there  was  a  team  of 
scientists  on  board,  they  began  to  see  that  if  they  just  acquired 
DNAX,  they  would  have  an  instant  presence  in  the  field  of 
immunology  research.   And  so,  after  a  few  discussions,  I  went  East 
with  Alex  to  make  a  presentation  to  the  scientific  board  of 
Schering.   We  thought  they  were  just  going  to  make  an  investment, 
keep  DNAX  separate.   But  then  Alex  came  back  and  said  their 
proposal  was  to  acquire  the  whole  thing.   And,  I  can  tell  you 
there  was  a  lot  of  discussion  and  resistance. 

People  thought  that  we  had  committed  to  building  something, 
creating  something  special.   We  had  this  sense  of  ownership,  and 
suddenly  we  were  selling  out.   But,  Alex  was  astute  and  recognized 
that  in  fact  all  the  value  that  DNAX  had  would  grow.   Because,  if 
Schering  Plough  and  its  fortunes  were  down,  and  they  ended  up,  we 
would  all  benefit  from  that.   So,  we  bought  into  it. 


158 
Scientists'  Initial  Reluctance 


Hughes:   What  about  the  effect  of  the  poor  image  research  in  the 
pharmaceutical  industry  had  in  the  academic  world? 

Berg:    Oh,  there's  no  question  that  existed.   That  was  part  of  the  thing. 
We  didn't  respect  their  science.   Frankly,  not  many  of  us  even 
knew  anything  about  their  science.  We  didn't  know  that  they  had 
not  been  a  very  successful  pharmaceutical  company  in  generating 
research,  in  generating  products.   They  were  just  some  big  enemy 
out  there,  part  of  the  pharmaceutical  industry  which  none  of  us 
had  a  lot  respect  for. 

But,  I  think  the  principle  thing  is,  or  at  least  my 
recollection  is,  we  felt  like  we  were  selling  out,  that  we  had 
started  this  [company]  with  a  grand  idea  that  we  were  going  to 
create  the  next  Syntex,  or  something  like  that.   We  had 
established  certain  criteria  that  DNAX  had  to  meet:  we  had  to 
have  a  very  academic  environment,  that  people  were  free  to 
publish,  that  people  would  be  able  to  talk  about  their  work.   All 
the  kinds  of  things  that  were  anti-traditional  to  pharmaceutical 
company  style,  we  were  rejecting.  And,  we  said  DNAX  is  going  to 
be  an  open  environment-- 

*f 

Berg:    --so  that  the  scientist  would  not  become  invisible  to  the  academic 
community  and  they  would  be  able  to  move  freely  back  into 
academia.   If  you  were  successful,  you  would  be  known.   You  would 
be  publishing  and  so  on.   So,  we  were  very  concerned  that  through 
this  acquisition,  all  of  what  we  promised  these  people  and  the 
kind  of  style  we  were  hoping  to  achieve  would  be  lost,  and  that 
Schering  would  not  honor  that  goal.   But  Alex,  I  think,  was  hugely 
successful  in  persuading  them  to  leave  DNAX  as  an  independent 
research  institute,  to  live  by  the  parameters  we  had  created  for 
its  lifestyle,  culture,  and  everything  that  we  had  told  people  was 
going  to  continue. 

Hughes:   If  he  had  not  been  successful,  do  you  think  you  would  have  stuck 
with  it? 

Berg:    Well,  I  think  one  would  have  had  to  see  just  how  offensive  it 
could  have  become. 


159 
J.  Allan  Waitz,  DNAX  President  and  CEO 

Berg:    In  fact,  we  came  very  close  to  rejecting  the  arrangement,  because 
once  the  deal  had  been  consummated,  we  were  sent  a  person  from 
Schering-Plough  to  become  the  president.  We  thought,  this  was  the 
beginning  of  the  end;  here  was  a  guy  who  was  going  to  instill  his 
culture,  their  culture  on  us.   But  it  turned  out,  he  was  totally 
swept  away  by  the  way  we  were,  and  Al  Waitz  became  the  great 
champion  to  the  point  of  offending  his  Schering-Plough  colleagues 
and  masters  by  how  he  defended  our  way  of  life,  and  the  way  we 
were  doing  it,  and  what  we  were  doing,  and  so  on. 

Hughes:   I  remember  from  Arthur's  book  the  comment  someone  at  Schering- 
Plough  made  to  Dr.  Waitz,  "Are  you  working  for  them  or  for  us?"1 
[laughter] 

Berg:    Al  was  terrific.   He  really  got  caught  up  in  California,  and  our 
style,  and  the  way  we  worked.   He  had  a  lot  of  influence  at 
Schering-Plough,  because  he  had  been  a  leader  in  their  interferon 
project.   He  had  worked  with  the  CEO,  [Robert  P.]  Luciano,  and  so 
Luciano  respected  his  views.  Al  protected  us  against  a  lot  of 
other  people. 


Tensions  between  DNAX  and  Schering-Plough 


Berg:     As  you  might  imagine,  there  was  a  lot  of  jealousy  because  we  were 
allowed  to  work  in  one  way  on  the  things  we  wanted,  and  people 
there  were  required  to  work  on  company  projects.  And  then,  the 
quality  of  the  science  in  the  two  places  was  a  mismatch.   I  mean, 
there's  no  question,  Schering-Plough  was  the  pits,  and  people  we 
had  at  DNAX  were  leading  people.   When  DNAX  reported  what  they 
were  doing  or  tried  to  get  collaborations  going,  they  lost  respect 
for  the  people  at  Schering-Plough.   DNAX  scientists  didn't  think 
Schering-Plough  scientists  were  quite  up  to  being  able  to  follow 
them.   So  I  think  in  the  beginning  there  was  a  lot  of  tension. 

Hughes:   Another  source  of  tension,  I  should  think,  would  have  been 

Schering-Plough' s  rather  ambivalent,  or  maybe  even  negative, 
experience  with  Biogen.   Remember  that? 


1  Arthur  Kornberg.   The  Golden  Helix;  Inside  Biotech  Ventures.   Sausalito, 
CA:  University  Science  Books,  1995. 


160 


Berg:    Yes.   Their  financial  deal  was,  they  made  an  investment  in  Biogen, 
and  in  return  they  got  the  complete  license  for  the  use  of  alpha 
interferon.   That  was  what  they  bought;  they  bought  a  product.   In 
order  to  buy  a  product,  they  had  to  make  their  investment.   I 
think  they  had  a  10  percent  ownership.  Al  Waitz  was  on  their 
board.   Hugh  d'Andrade  was  on  their  board.   I  don't  think 
Schering-Plough  was  disillusioned  with  Biogen  until  quite  a  bit 
later.   And  in  the  end,  they  got  a  big  financial  return.   Not  only 
do  they  have  a  product  which  today  is  a  three-to-four-hundred- 
million-dollar-a-year  product,  but  in  addition,  their  investment 
in  the  ownership  of  stock  went  up.   So  they  did  quite  well. 

But  the  DNAX  thing  was  really  more  of  a  strategic 

investment,  that  is,  if  they  had  decided  that  immunology  was  going 
to  be  the  core  of  their  research  program,  and  the  entree  into 
infectious  disease,  cancer,  and  things  of  that  sort,  they  needed  a 
strong  core. 

Hughes:   Who  were  the  main  competitors  in  that  area?   Presumably  other 
pharmaceutical  companies  had  also  targeted  immunology? 

Berg:    Oh,  yes.   I  think  immunology  was  seen  as  a  central  player.   I 

don't  know  who  else  staked  their  strategic  future  to  the  extent 
that  Schering  did.   Big  companies  probably  didn't  have  to.   But, 
Schering-Plough  was  really  groping.   They  didn't  have  anything, 
and  the  market  knew  that . 

Hughes:   Well,  as  you  know,  once  the  Schering-Plough  acquisition  occurred, 
there  was  a  shift  in  the  research  agenda. 

Berg:  Yes. 

Hughes:  Would  you  like  to  talk  about  those  decisions? 

Berg:  It's  twelve  o'clock. 

Hughes:  Oh,  that's  what  that  means? 

Berg:  Yes,  my  little  beeper  beeps. 

Hughes:  It  tells  you  when  you're  hungry?   [laughter] 

Berg:    No,  it's  there  because  this  watch  has  to  have  an  alarm  setting. 

And  so  I  conveniently  put  it  at  noon  because  I'm  up.   If  I  put  it 
at  seven  o'clock  in  the  morning,  I  don't  want  to  wake  up.   So,  I 
had  to  find  some  time  [to  program  it  to],  and  it  always  evokes 
some  comment,  "Oh,  is  that  my  time  up,"  or  something  like  that. 
But  actually,  I  booked  us  for  lunch,  like  I  did  last  time, 
[interruption] 


161 


Maxine  Singer  was  invited  to  the  meeting  that  we  had  at  MIT 
where  we  drafted  the  "moratorium",  or  "Berg",  letter.   But  she  was 
taking  her  kids  to  Disney  World  in  Florida  and  decided  that  was 
more  important  than  being  at  that  meeting.   But  when  we  came  out 
with  that  letter  and  were  planning  to  have  the  Asilomar  meeting,  I 
certainly  her  to  be  a  participant  on  the  organizing  committee. 

Her  husband,  Dan,  was  at  the  Asilomar  meeting  and  was  very 
influential.  As  the  history  goes,  the  lawyers  opened  our  eyes  to 
our  collective  responsibility,  obligation,  and  vulnerability  to 
being  sued,  and  so  on  and  so  forth.   Dan  was  the  one  who  organized 
the  discussion  that  evening  by  the  lawyers  and  helped  to  identify 
the  people  to  invite,  all  of  whom  were  very  influential  in  the 
outcome. 


ALZA  and  DNAX 


Hughes:   I  have  a  news  release  of  March,  1981,'  which  was  before  Schering- 
Plough  acquired  DNAX  the  next  year.   Zaffaroni  announced  that 
DNAX's  strategy  was  "unique  in  combining  three  technologies- 
genetic  engineering,  immunobiology ,  and  drug  delivery  systems." 
Now,  the  drug  delivery  systems  was  a  Zaffaroni  connection,  was  it 
not? 

Berg:    Well,  one  of  the  investors  in  DNAX  was  ALZA;  $600,000  worth,  1 
think,  out  of  the  $4.5  million  that  bankrolled  the  company. 
Zaffaroni's  vision  was  that  we  could  make  proteins  through  genetic 
engineering,  and  that  ALZA  was  going  to  develop  delivery  devices 
for  delivering  proteins.   Remember,  delivering  proteins  across 
membranes  is  not  a  trivial  job.   And,  all  of  the  delivery  devices 
that  they  had  made  until  then  were  delivering  small  molecules. 

He  foresaw  that  if  you  made  therapeutic  proteins,  delivering 
them  at  known  sites,  known  rates  of  release  would  be  important. 
And  so  ALZA  was  supposed  to  be  part  of  this  kind  of  consortium 
that  would  design  delivery  devices  for  proteins.   They  still 
haven't  done  it.   Delivering  proteins  is  still  a  big,  big 
challenge. 


1  [News  release],  March  9,  1981.  (Papers  in  the  possession  of  Arthur 
Kornberg  concerning  DNAX  and  his  book  The  Golden  Helix.  "Helix"  carton) 


162 
DNAX's  Original  Product  Goal 

Berg:    The  whole  thing  that  we  were  focused  on  was  how  to  use  the 
recombinant  DNA  technique  to  actually  make  designed 
immunoglobulins  with  very  special  properties.   We  talked  about 
being  able  to  make  enormous  columns,  if  you  will,  of  protein 
molecules,  mobilized  on  supports  through  which  you  could  pour 
crude  mining  extracts.  And  they  would  extract  the  gold,  or  they 
would  extract  some  very,  very  low  abundance  material  because 
antibodies  have  an  incredibly  high  affinity.  And  you  could  use 
genetic  engineering  to  modify  the  combining  sites  of  natural 
proteins  to  increase  their  binding  specifity  and  so  on.   Alex 
actually  got  a  patent.   It  was  for  high-affinity  binding  sites,  or 
something  like  that.   I  forget  what  it  was  called. 

[perusing  documents]  Dynapol  was  the  other  company  [of 
Zaffaroni's]  that  Arthur  was  involved  with.   It  was  a  spin-off 
from  ALZA. 

Hughes:   Is  that  the  company  you  were  referring  to? 

Berg:    Yes.   I  think  Arthur  says  he  lost  his  shirt  on  that.   [laughter] 

One  of  the  important  things  that  emerged  after  the 
acquisition,  which  really  helped  delay  some  of  our  fears  about 
what  was  going  to  happen,  was  that  DNAX  was  to  be  governed  by  a 
policy  board.   The  policy  board  was  to  have  me,  Charlie,  Arthur, 
Zaffaroni,  and  three  representatives  from  Schering-Plough,  and 
that  balanced  how  things  were  to  be  decided.   These  policy  board 
meetings  would  be  held  here  in  Palo  Alto.   Like  a  board  of 
directors  of  a  company,  it  would  act  as  a  board  of  directors  for 
this  common  joint  venture,  and  that,  again,  helped  make  us  feel 
that  Schering-Plough  was  willing  to  make  concessions.   Then,  there 
were  reassuring  statements  about  East  is  East  and  West  is  West, 
and  we're  not  going  to  try  to  blend  the  two.   The  personnel 
policies  were  different  for  DNAX  than  they  were  for  Schering- 
Plough  employees.   So  they  clearly  recognized  that  there  was 
something  special  about  DNAX  and  it  had  to  be  nurtured. 

I  shouldn't  say  they  realized.   They  were  taught,  totally 
encouraged  to  believe  that.   [laughter]   At  every  meeting, 
Zaffaroni,  Arthur,  and  I  would  beat  on  them  about  the  very  special 
quality  of  DNAX  and  the  risk  of  losing  it.   In  fact,  I  remember 
when  Alex  was  trying  to  talk  us  into  going  for  this  merger,  he 
said,  "What  have  you  guys  got  to  risk?  Let's  assume  that  they 
come  in  here  and  they  transform  DNAX  into  Schering-Plough  West. 
You  guys  leave,  what  have  they  bought?  They  bought  you.   That's 
what  the  value  is.   There's  no  product;  there's  no  anything;  they 


163 


bought  you.   So  in  order  to  make  their  investment  worthwhile,  they 
have  to  enlist  your  compliance."  And  we  realized  that  was  true. 


The  Shift  to  T-cells 


Hughes:   There  was  a  shift  in  research  direction  after  the  Schering-Plough 
acquisition.   It  was  not  going  to  be  antibodies,  but  interleukins . 

Berg:    Well,  what  happened  is  in  this  strategic  review  they  had,  the  part 
of  immunology  which  Schering-Plough  got  sold  on  was  T-cells. 

Hughes:   Who  was  pushing  T-cells? 

Berg:    This  was  a  guy  named  Harvey  Cantor,  who  was  a  professor  at 
Harvard,  who  was  a  good  immunologist.   He  had  been  on  this 
strategic  advisory  board,  and  he  had  persuaded  them  and  the  rest 
of  the  people  that  T-cells  were  the  crux  of  the  immune  system,  as 
it  was  understood  in  1980.   They  are  the  cells  that  sense  antigen 
first,  and  they  elicit  the  rest  of  the  immune  response  by 
secreting  cytokines,  interleukins  as  you  call  them.   He  had 
already  done  some  studies  to  show  that  a  certain  cell  line,  when 
stimulated,  produced  all  kinds  of  cytokines. 

So  when  Schering  discussed  the  acquisition  of  DNAX,  they 
said  they  were  not  interested  in  antibody  production;  they  were 
not  interested  in  making  these  mickey  mouse  little  things  that 
could  be  injected  to  cure  overdosing  on  Digoxin.   They  would  be 
interested  if  we  would  work  on  T-cells.   Nobody  felt  totally 
wedded  to  the  antibody  project.   That  was  always  viewed  as  being 
just  to  get  us  in  the  door,  just  to  get  the  company  started. 

Hughes:   Not  much  research  on  antibodies  was  already  done? 

Berg:    It  had  been  going  along  for  a  bit.   It  clearly  wasn't  succeeding, 
and  we  probably  would  have  been  confronted  with  making  some  new 
choices.   But  when  they  said  T-cells  in  trying  to  identify  these 
cytokines,  it  was  clear  we  could  translate  the  same  technology, 
that  is,  cloning  cDNAs.   Within  an  instant,  Kenichi  Arai  and  those 
guys  took  the  cell  lines  from  Harvey  Cantor  and  started  cloning 
out  cDNAs  for  all  kinds  of  things. 


164 


The  Expression  Vector  Technique 

Berg:    One  of  the  major  things  they  had  adopted  was  a  technique  Okayama 
and  I  developed  called  an  expression  vector.   Expression  vector 
means  that  when  you  clone  the  cDNA  into  the  vector,  it's  already 
in  place  to  be  expressed  to  make  the  protein.   Whereas  with  normal 
cloning,  you  just  clone  it  into  a  plasmid.   After  you've  cloned  it 
you  try  to  recognize  it  by  its  sequence.   But  if  you  don't  know 
anything  about  the  gene  sequence,  and  you're  looking  for  a  gene 
that  makes  a  kind  of  protein  that  has  a  biological  activity,  you 
want  a  system  where  you  make  the  proteins  in  the  cloning 
operation.   We  had  developed  this  expression  cloning  system,  and 
that  was  probably  the  single  most  important  contribution  to  the 
success  of  DNAX.   DNAX  cloned  all  kinds  of  cytokines. 

Hughes:   DNAX  had  this  expression  system  when  other  companies  did  not? 

Berg:    Other  companies  did  not.   But  very  quickly  others  recognized  that 
that  was  the  way  to  go.   So  they  developed  variants,  all  of  which 
had  the  same  capability  so  that  when  you  cloned  the  sequence,  it 
was  in  a  position  behind  the  promoter  so  it  could  be  expressed  in 
animal  cells  or  in  any  system  you  wanted. 

The  company  Genetics  Institute  developed  an  expression 
system;  actually  it  was  a  postdoc,  Steve  XX  [Berg  can't  recall  the 
full  name]  from  my  lab  who  had  been  there  when  Okayama  developed 
the  expression  cloning  system,  who  then  went  to  Genetics  Institute 
and  developed  a  comparable  promoter  using  adenovirus.   We  were 
using  plasmid  systems.   Essentially,  it  took  advantage  of  the  fact 
that  you  could  clone  full-length  cDNAs,  and  when  they're  cloned, 
they're  put  into  a  vector  that  expresses  them.   So  DNAX  was  a  new 
gene  a  month.   I  mean,  it  was  really  booming  because  it  had 
developed  a  very  good  way  of  cloning  rare  things  that  had  never 
been  known  before. 


Recruitment  of  Scientists 


Hughes:   Whom  did  DNAX  recruit? 

Berg:    There  was  the  core  group:  Kenichi  Arai  because  he  came  back  from 
Japan,  Gerard  Zurowski  came  back  from  Australia,  Kevin  Moore  from 
Lee  Hood's  lab,  a  few  others.   DNAX  tried  to  recruit  Okayama, 
because  he  was  a  whiz .   But  he  came  from  a  Japanese  background 
where  he  thought  he  would  be  unlikely  to  ever  ^et  a  job  in 
academia  if  he  went  into  industry. 


165 

Hughes:   It  probably  would  have  been  true  at  that  time,  would  it  not? 

Berg:    At  that  time,  it  might  have  been  true.   Today,  it's  not  at  all 

true.   Kenichi  Arai  went  back  as  the  professor  of  biochemistry  in 
the  Institute  for  Science  and  Technology  at  Tokyo  University,  and 
Okayama  went  from  being  the  professor  in  Osaka  to  Tokyo 
University. 

Hughes:   It  sounds,  from  the  way  you've  just  described  it,  that  the  way 
these  people  came  to  DNAX  was  largely  through  personal 
connections . 

Berg:    That's  right.  And  a  lot  of  selling—selling  in  the  terms  that 

this  was  going  to  be  a  congenial,  exciting,  productive  environment 
that  would  be  every  bit  as  academic  as  any  academic  position  would 
be.  They  would  be  paid  well  and  have  a  stake  in  the  future  of  the 
company,  and  they  were  free  to  publish  and  talk  about  their  stuff. 


Research  Freedom 


Hughes:   Also,  and  this  really  surprised  me,  DNAX  scientists  were  given  the 
opportunity  to  pursue  their  own  research. 

Berg:    That's  right.   Within  certain  bounds.   Somebody  wanting  to  work  on 
photosynthesis  would  not  come  to  DNAX. 

Hughes:   There  was  somebody  at  DNAX  working  on  photosynthesis. 
Berg:    There  was,  and  Gerard  had  been  doing  it  before. 

Everybody  was  assured  that  they  had  a  certain  fraction  of 
time,  20,  30,  40  percent  of  the  time,  that  they  could  do  whatever 
they  wanted.   And  their  other  research  was  to  relate  to  the 
principle  theme  of  DNAX.   But  the  principle  theme  was  very  broad. 
It  was  molecular  immunology  and  subsequently  became  molecular 
immunology  and  cell  growth  control.  Within  those  boundaries, 
people  could  do  almost  anything. 

Gerard  Zurowski,  who  was  working  in  Australia,  was  doing 
some  plant  work.   He  came  here  and  carried  on  the  plant  work  for  a 
while .   But  he  got  so  caught  up  in  the  immunology  part  of  it  and 
the  plant  work  eventually  went  by  the  wayside,  and  that  happened 
for  most  people.  The  work  was  so  exciting  and  successful  that 
most  people  got  drawn  into  it  and  left  behind  whatever  thing  they 
had  in  mind  before. 


166 


Hughes:   I  spoke  of  the  cloning  gold  rush.   I  saw  some  memos  that  stated 
that  after  a  few  years,  there  was  a  lull  at  DNAX  and  other 
companies  rushed  in  and  actually  took  the  plums.1  What  was  your 
feeling  at  that  point? 

Berg:    Well,  my  recollection  is,  the  first  plateau  was  all  these 

cytokines  were  being  mined  successfully  by  Immunex,  which  is 
another  company,  and  Genetics  Institute.   There  were  a  number  of 
times  that  we  won,  a  number  of  times  they  won.   That  is,  they  got 
to  the  goal  first  even  though  we  were  all  competing  for  the  same 
thing . 


Cloning  Cytokine  Receptors 

Berg:    But  then  we  came  to  the  realization  that  the  next  big  direction 
was  to  understand  the  receptors  to  which  these  cytokines  bind. 
The  way  cytokines  act  is  they  bind  to  receptors  that  are  on  the 
cell,  and  then  there  is  a  signal  transduction;  something  gets 
passed  on  to  the  nucleus,  which  then  triggers  the  cell  to  do 
something  to  respond  to  that  signal.   What  we  began  to  realize  is 
that  just  knowing  the  first  part  of  it,  what  are  the  things  that 
you  put  out  as  hormones,  wasn't  enough;  we  had  to  understand  how 
the  cell  responded  to  those  hormones,  those  signals.   That's  the 
crux  of  the  immune  response. 

So,  we  had  to  pursue  new  goals.   One  of  those  new  goals  was 
to  start  cloning  the  receptors  specifically  for  these  cytokines, 
and  ultimately  to  understand  the  pathway  by  which  a  signal 
transmitted  from  the  receptor  to  the  nucleus.  And  that,  today, 
constitutes  the  principle  mission  of  DNAX.  What  DNAX  had  as  an 
edge  was  a  very  special  skill  in  isolating  and  focusing  on  the 
rare  cells  of  the  immune  system.   In  other  words,  you  could  take 
T-cells,  but  then  there  are  subclasses  and  subclasses,  and  there 
are  different  kinds  of  B-cells  at  various  stages.  And  all  of  them 
have  unique  patterns  of  gene  expression.   So  if  you  can  begin  to 
enrich  for  certain  cell  types,  you  have  access  to  certain  kinds  of 
proteins  that  somebody  looking  at  the  bulk  won't  ever  find.   And 
so  that  has  been  the  DNAX's  expertise. 

Today  DNAX  is  moving  along  working  with  cell  types  which 
probably  very  few  people  in  the  world  know  how  to  isolate,  grow, 
and  adapt.   DNAX  is  looking  for  gene  discovery  in  those  cells 


1  For  example,  see:   J.A.  Waitz  to  Policy  Board  Members,  December  29, 
1986.   (Arthur  Kornberg  personal  papers,  DNAX  1987-1988  [sic]) 


167 


under  the  assumption  that  if  you  identify  the  important  protein  in 
that  system  you  might  have  an  edge  into  a  particular  function, 
maybe  a  disease,  and  so  on. 


A  Long  Discovery  Phase 

Hughes:   Arthur  in  his  book  writes  about  the  congeniality  between  the 

people  at  DNAX  and  Schering-Plough.   But  I  nonetheless  suspect 
that  there  were  some  tensions.   Did  people  like  Luciano,  the  top 
guys,  appreciate  how  long  it  would  take  from  the  discovery  phase 
to  an  actual  product? 

Berg:    They  did.   They  told  us  right  in  the  beginning  that  they  did  not 
expect  to  see  any  products  from  DNAX  research  for  probably  the 
order  of  ten  to  twelve  years. 

Hughes:   How  could  they  have  known  that? 

Berg:    If  you  take  a  strategic  view  to  the  pharmaceutical  industry,  you 
have  to  think  long-term.   It  takes  a  long  time  from  even  the  most 
promising  discovery  in  the  lab  until  you  get  something  you  can 
sell.   And  many  things  fall  by  the  wayside.   So  they  knew  the 
game. 

Hughes:   From  past  experience. 

Berg:    Yes.   I  think  what  they  were  trying  to  do  was  to  take  the  pressure 
off  us.   They  wanted  us  to  do  cutting-edge  research.   They  wanted 
us  to  be  at  the  very  frontier  of  this  field,  with  the  confidence 
that  it  was  going  to  lead  to  commercial  value.   And  they  didn't 
want  us  to  feel  that  we  had  to  prove  tomorrow  that  we  had  made 
something. 


DNAX  Benefits  Schering-Plough 


Berg:    But  in  point  of  fact,  within  a  very  short  time,  Schering-Plough 
had  patents  on  a  lot  of  very  valuable  stuff. 

Hughes:   But  an  actual  product  wasn't  on  the  market  until  about  1990. 

Berg:    That's  right.   But  Schering  knew  that  they  had  things  in  the 
pipeline  that  were  potentially  very  valuable.   But  more 
importantly,  they  also  recognized  that  DNAX  scientists  had  made  an 


168 

incredible  reputation  and  inroads  in  the  field  of  immunology. 
They  were  now  looked  upon  as  leaders  of  the  field.   And  for  an 
organization  that  had  a  bad  image  in  the  research  field,  DNAX  was 
a  jewel  in  their  crown.   I  mean,  they  could  boast  that  the  number 
of  papers  published,  number  of  symposium  speakers,  and  by  almost 
every  criterion,  DNAX  was  one  of  the  leading  research  places  in 
immunology. 

DNAX  had  made  several  discoveries,  which  while  they  didn't 
have  commercial  rank,  had  radically  transformed  the  field  of 
immunology.   I  mean,  the  whole  idea  that  there  are  two  classes  of 
T-helper  cells,  each  secreting  unique  sets  of  cytokines  was  a 
bombshell.   And  to  this  day,  DNAX  is  cited  as  the  place  where 
these  discoveries  occurred.   Today,  DNAX  scientists  are  considered 
amongst  the  leading  ones  in  the  world. 

Hughes:   How  did  Schering-Plough  commercialize  this  very  prominent  research 
group  that  they  had  on  the  West  Coast?   How  did  it  help  them  sell 
their  products? 

Berg:    Well,  I  don't  think  it  helped  them  sell  their  products.   There  are 
two,  I  call  them,  intangibles. 


Berg:    There's  no  question  that  today  Schering-Plough  in  New  Jersey  is  a 
first  rank  research  organization,  whereas  when  DNAX  first  joined 
them,  it  was  pitiful.   So  there  has  been  a  transformation  in  their 
research  organization. 

Hughes:   How  did  that  happen? 

Berg:    Well,  in  large  part  because  they  recognized  the  disparity;  that 
things  coming  from  DNAX  couldn't  be  implemented  at  Schering- 
Plough.   People  at  Schering  didn't  fully  understand  them. 

Hughes:   So  Schering  began  to  attract  better  scientists? 

Berg:    Yes,  with  our  help.  We  helped  recruit  people.  We  have  been  very 
active  in  Schering-Plough' s  activities,  in  recruiting  leadership 
in  various  positions.   They  don't  hire  anybody  in  the  science 
organization  unless  we  approve  it. 

Hughes:   Is  that  by  contract  or  is  that  just  an  understanding? 

Berg:    I'll  call  it  dependence.  And  recognition.   Recognition  of  what  we 
contribute.   Nobody  is  hired  at  DNAX  without  each  of  us  having 
interviewed  them.   I  mean,  Arthur,  Charlie,  and  me.   We've  done  a 
lot  of  active  recruiting,  that  is,  persuading  people  who  are  on 


169 

the  fence  about  whether  they  want  to  go  to  academia  or  go  to 
industry. 

The  second  point  is,  DNAX  had  been  incredibly  efficient  in 
generating  new  kinds  of  cell  cultures,  which  then  could  be  used 
for  assaying  special  kinds  of  things.   These  were  very  valuable, 
and  all  of  these  were  transferred  to  Schering.   So  Schering  got 
technology,  got  materials,  potential  drugs,  some  of  which  are 
still  in  clinical  trial,  and  a  turnover  in  their  own 
establishment.   So,  they've  been  transformed.  And  I  think  they 
rightly  give  DNAX  a  lot  of  credit  for  having  catalyzed  it,  aided 
it.   You  could  say,  well,  the  way  commercial  companies  judge  value 
is  not  always  the  way  we  judge  it.  We  think  of  it  in  terms  of 
big,  big  accomplishments.   But  the  transformation  of  a  nearly 
moribund  scientific  organization  on  which  a  pharmaceutical  company 
has  to  depend  into  one  which  is  now  recognized  and  can  recruit 
very  easily  is  an  important  accomplishment. 

Second,  their  investment  looks  very  good  because,  for  not  a 
lot  of  money,  about  $28  million  at  the  time  of  the  acquisition, 
they  bought  a  world-class  research  organization.  And  they  got  a 
lot  of--.   Which  word  do  I  want? 

Hughes:   Kudos? 

Berg:    Yes.  --for  having  done  it.   In  fact,  Arthur  wrote  this  book 

[Golden  Helix]  because  he  was  told  that  DNAX  was  used  as  a  case 
study  in  business  schools.  People  said,  gee,  somebody  ought  to 
write  this  all  down. 


Arrival  at  Stanford.  1959 
Advance  Preparation 


Hughes:   In  our  first  interview,  you  said  that  you  wanted  to  talk,  and  we 
did  not,  about  your  earliest  experiences  at  Stanford, 
[interruption] 

Berg:    You  have  enough  material  about  how  we  got  recruited  to  come  to 
Stanford. 

Hughes:   Yes,  I  agree. 


170 

Berg:     So  we  had  two  years  lead  time  to  prepare  for  it,  and  that  two 

years  was  one  of  active  designing  the  building  in  which  we  were 
going  to  move,  its  interior  wasn't  yet  designed,  and  preparing  for 
what  we  hoped  was  going  to  be  a  totally  novel  way  of  teaching 
biochemistry.   Remember  that  the  group  that  moved  here  from 
Washington  University  were  technically  all  microbiologists;  we 
were  in  the  microbiology  department.  We  considered  ourselves  all 
biochemists,  and  in  some  ways  we  were  parading  under  false 
pretenses . 

Hughes:   Standards. 

Berg:     The  move  here  was  looked  to  going  to  do  something  really  novel  and 
exciting  in  biochemistry.   During  those  two  years  we  helped 
develop  a  teaching  program  that  was  really  remarkable  and  lasted 
for  at  least  five  to  eight  years  in  terms  of  being  the  most 
popular  course  at  the  medical  school. 


An  Unfinished  Science  Building 

Berg:    In  June  of  1959,  we  were  supposed  to  come  here  from  St.  Louis  and 
move  into  the  new  building.  When  we  got  here,  the  building  wasn't 
completed.   We  were  told  that  it  was  on  the  way,  and  we  should 
just  wait.   So  we  camped  out,  in  a  sense.   We  had  a  hut  where  the 
secretary  sat  who  came  with  us.   And  we  would  come  in  every  day 
and  say,  "Any  news?  Any  news?"  And,  we  would  hear,  "No,  they 
haven't  done  anything." 

So,  eventually,  we  got  so  frustrated  that  Arthur,  who  had  a 
lot  of  clout,  went  to  the  administration  and  said  he  was  outraged, 
furious,  and  so  on  and  so  forth.  And  so  what  they  did  was  gather 
all  the  workmen  that  were  working  all  over  the  medical  center,  in 
the  hospital  and  everyone  out  there,  onto  the  third  floor  of  the 
medical  school  building.  And  they  finished  us  up  in  a  very  short 
period  of  time. 

We  had  these  moving  vans  that  had  transported  all  our  lab 
equipment  and  supplies,  and  we  moved  right  in.   We  moved  in 
probably  by  sometime  in  July,  and  by  the  middle  of  August 
everybody  was  doing  experiments. 


171 


Settling  In 


Hughes:   And  teaching? 

Berg:    We  started  teaching  again  in  September.   We  had  a  new  course.   We 
were  all  teaching  in  new  areas.   But  the  research  was  going  on. 
We  had  every  barrel,  every  box  marked,  ticketed  almost  exactly  to 
which  drawer  everything  was  supposed  to  go  in.   We  had  worked  very 
hard  to  sterilize  and  wrap  all  the  stuff  we  wanted  to  take.   When 
we  came,  we  had  a  carpenter;  he  built  the  dividers  and  drawers. 
It  went  from  the  barrel  right  into  the  drawer. 

The  only  glitch  we  had  was  that  there  were  a  lot  of  air 
vents  and  things  built  in,  and  we  couldn't  get  certain  instruments 
onto  the  table  because  these  things  were  sticking  out.   So  we 
unscrewed  them  and  capped  them.   But  the  building  had  never  been 
officially  tested.   They  put  pressure  on  all  the  air  lines  or  the 
gas  lines  to  see  that  they  don't  leak.  Well,  of  course,  they 
leaked  like  a  sieve  because,  what  we  had  put  in  was  not  standard, 
[laughter]   So  we  got  the  fire  department  down  our  back.   There 
was  a  little  flap. 

Hughes:   They  knew  that  biochemists  were  in  town.   [laughter] 

Berg:    Yes,  and  working  very  quickly.   It  was  really  an  amazing  move. 

Hughes:   You  had  been  orchestrating  it  for  almost  two  years  from  St.  Louis, 
from  what  you  said. 

Berg:    Everything  was  planned  down  to  a  T.   We  brought  with  us 

secretaries,  technicians,  students,  even  shop  people.   And  so  we 
had  everybody  ready  to  transform  the  floor  into  a  working  unit. 


The  Stanford  Department  of  Genetics 


Lederberg's  Arrival  at  Stanford 


Hughes:   I  got  the  impression  from  going  through  the  archive  that  a 

collaboration  or  a  partnership,  whatever  you  want  to  call  it,  with 
the  Department  of  Genetics  headed  by  Lederberg  was  really  on 
people's  minds.   And  yet,  from  what  I  can  tell,  it  did  not  really 
materialize.  Am  I  right? 


172 

Berg:    Yes,  to  a  certain  extent.   Josh  Lederberg  was  in  the  field  of 

genetics,  probably  one  of  the  shining  bright  stars.   He  had  been 
invited  to  come  to  Stanford  and  turned  it  down.   But  when  Kornberg 
and  we  accepted  to  come,  I  think  it  might  have  been  Kornberg  who 
contacted  him  and  said,  "Think  again.  We're  coming,  and  I  think 
that  between  your  genetics  and  our  biochemistry,  we  can  really  be 
a  powerhouse."  And  Josh  changed  his  mind. 

Now,  unfortunately,  when  Josh  changed  his  mind,  there  was  no 
space,  because  there  hadn't  been  any  program  to  have  a  genetics 
department.   So  we  gave  him  space  in  our  department. 

Hughes:   There  had  been  no  genetics  department? 

Berg:    There  had  been  no  genetics  department.   In  fact,  I  think  Stanford 
was  the  first  medical  school  to  actually  create  a  genetics 
department . 

Hughes:   Is  that  so? 
Berg:     Yes,  1959. 

Hughes:   So,  it  was  the  Department  of  Genetics  in  the  Department  of 
Biochemistry? 

Berg:    Well,  it  was  the  Department  of  Genetics  autonomous  in  every  way. 
But  the  space  that  they  used  was  in  biochemistry  space. 


Stanley  Cohen's  Associations  with  the  Biochemistry 
Department 


Hughes:  Because  Genetics  and  Biochemistry  were  in  more  or  less  the  same 
place,  it  was  easy  for  people  like  Stan  Cohen  and  Sgaramella  to 
participate  in  biochemistry  seminars.  Is  that  right? 

Berg:    That's  right.   Only  Stan  never  was  really  located  in  that  space, 
because  when  Stan  came  to  Stanford  in  1968  he  was  in  the 
Department  of  Medicine.   He  was  recruited  to  become  the  director 
of  the  Division  of  Clinical  Pharmacology.   So  he  had  space 
somewhere  else. 

I  knew  him  before  because  he  came  from  a  lab  of  one  of  my 
closest  friends,  Jerry  Hurwitz  at  Albert  Einstein  College  of 
Medicine.   I  knew  about  Stan  and  when  he  came,  I  certainly  had  an 
open  feeling  for  him.   He  then  hit  on  plasmids  as  the  thing  to 
study  in  the  area  of  drug  resistance,  which  was  clinical 


173 

pharmacology.   When  we  found  that  the  enzyme  EcoRl  would  make 
cohesive  ends,  and  Mort  Mandel  discovered  how  to  incorporate  DNA 
into  cells  by  giving  them  calcium  shock.   Stan  started  hanging  out 
in  Biochemistry  because  he  saw  that  we  were  working  now  with 
plasmids.  My  student  Janet  Mertz  was  doing  all  these  experiments, 
and  she  taught  Stan  how  to  do  these  transformations. 

Hughes:   So,  up  until  then,  he  hadn't  been  around  very  much? 

Berg:    No,  not  at  all.   Walter  Bodmer,  who  is  now  a  distinguished 

professor  of  genetics  at  Oxford,  interacted  a  good  deal  with  our 
department.   He  and  Josh  formed  the  core  of  the  genetics 
department.   They  had  some  postdocs,  one  of  whom  went  on  to  become 
the  head  of  the  Eliza  Hall  Institute,  Gus  Nossal.   Nossal,  Bodmer 
and  Leonard  Hertzenberg  were  on  our  floor. 


Lederberg  and  Space  Biology 


Berg:    Josh  was  somewhat  aloof  and  had  very  little  to  do  with  the 

research.   In  fact,  at  that  time  Josh  was  involved  with  planetary 
biology.   He  was  learning  about  rockets  and  space  biology  and  so 
on.   So  he  really  tuned  out. 

Hughes:   Was  that  soon  after  he  arrived? 

Berg:    Yes,  because  I  remember,  in  1961  I  went  to  the  International 

Congress  of  Biochemistry  in  Moscow.   Josh  was  already  actively 
involved  in  space  biology  because  he  enlisted  me  to  sit  in  for  him 
at  this  meeting  in  Moscow  with  other  space  biologists.   I  didn't 
know  anything  about  it.   But  anyway,  he  was  very  heavy  into  it. 

Josh  told  me,  he  denies  it  today,  but  I  remember  very 
clearly.   He  used  to  come  walking  by  my  lab  on  his  way  out, 
because  his  office  was  what  became  my  office  when  I  became 
chairman.   He  was  on  my  corridor.   He  often  would  stop  in  with  his 
arms  full  of  books  on  planets,  cosmology,  astronomy,  and  rockets. 
He  would  come  back  the  next  morning  with  the  same  books  already 
digested.   He  was  remarkable. 

Anyway,  he  said  that  he  left  what  he  had  been  doing  before 
in  genetics,  in  part  because  he  realized--  The  structure  of  DNA 
was  published  in  1953.   From  '53  to  '59,  Josh  was  skeptical  of  the 
notion  that  DNA  could  explain  the  gene's  properties.   He  argued 
that  genetics  couldn't  be  explained  by  this  molecule;  there  were 
too  many  complicating  features  about  genetics  that  couldn't  be 
explained  by  this  simple  molecular  structure.   Josh,  I  th:.nk,  was 


174 


one  of  the  last  hold-outs,  but  by  the  time  he  came  to  Stanford,  he 
was  convince  that  it  was  correct.   Nevertheless,  I  think  Josh 
recognized  that  he  was  out  of  it  because  he  was  never  molecularly 
oriented;  he  was  largely  a  classical  geneticist.   And  now  genetics 
had  become  molecular,  and  he  wasn't  into  it.   Walter  Bodmer,  along 
with  Ganesan,  a  student  of  Josh's,  was  doing  transformation; 
Nossal  was  doing  immunology,  and  Josh  had  to  find  something  new. 
Space  biology  was  sort  of  blossoming  as  a  possibility,  and  he  just 
went  right  for  that.   Josh  says  today  that  he  doesn't  remember 
ever  saying  that.   But  I  remember  very  clearly  because  I  was 
really  astonished  to  see  how  somebody  who  was  as  extraordinary  as 
he  is  eventually  felt  that  he  had  been  eclipsed. 


Faculty  and  Tenor  of  the  Genetics  Department 


Hughes:   What  about  the  other  people  in  Genetics?  Was  there  much 
interchange  with  Biochemistry? 

Berg:     No.   There  was  Indian  fellow  named  Ganesan.   He  was  a  graduate 
student,  and  he  worked  with  Bodmer  on  DNA  transformation.   Josh 
brought  Luca  Cavalli-Sforza,  who  was  a  human  geneticist,  to 
Stanford.   A  wonderful  man  who  had  had  a  strong  history  in 
microbial  genetics  but  now  had  gone  into  human  genetics. 

It  was  an  interesting  culture.   Josh's  department  was  very 
different  than  Biochemistry.   Biochemistry  was  extremely 
interactive.   Josh's  department  was,  you're  on  your  own. 
Everybody  was  on  their  own.   There  were  very  few  faculty  meetings 
It  was  a  non-department.   Josh  was  involved  in  traveling  around 
the  globe  doing  his  things.   People  were  doing  their  own  things. 


Minimal  Interaction  between  Biochemistry  and  Genetics 


Berg:    I  think  Arthur  would  probably  say,  and  I  would  too,  that  a 
coalescence  between  Genetics  and  Biochemistry  never  really 
happened.   It  didn't  happen  in  the  teaching;  it  didn't  happen  in 
research.   There  was  virtually  no  collaborations  that  were 
established.  And  there  was  no  real  intellectual  interactions.   We 
knew  them  and  we  knew  what  they  were  doing.   I  think  the  closest 
it  came  was  when  Stan  started  coming  up  to  the  department  to 
interact,  with  Mort  Mandel,  who  was  a  visitor  in  Dale  Kaiser's 
lab,  and  with  my  graduate  student,  Janet  Mertz. 


Hughes:   How  about  Sgaramella? 

Berg:    Sgaramella  was  also  a  postdoc  in  genetics.   He  had  come  from 

Khorana's  lab.   He  came  to  my  group  meetings,  and  I'm  sure  he  came 
to  biochemistry  department  seminars,  but  I  don't  remember  that  as 
well.   But  I  never  saw  him  as  a  strong  element  or  a  strong  part  of 
the  genetics  department.   He  was  a  transient,  who  had  already  come 
with  a  problem.   I  mean,  the  problem  that  he  worked  on  in 
Khorana's  lab  was  this  discovery  that  T4  ligase  would  join  blunt- 
ended  molecules.   He  certainly  came  to  our  group  meetings  and  he 
participated  in  the  discussions.   I'm  sure  he  must  have  given  a 
presentation  of  his  own  work,  but  I  don't  remember  it. 


Biochemistry's  Policy  on  Joint  Appointments 


Hughes:   So  it  wasn't  a  rich  exchange  back  and  forth? 

Berg:    No.   But  you  also  have  to  remember  that  Biochemistry  was  somewhat 
aloof.   Biochemistry  itself  was  quite  snooty;  it  didn't  interact 
with  anybody.   It  had  a  policy:  it  would  not  offer  joint 
appointments  to  anybody,  whereas  lots  of  other  places  would  help 
in  the  recruitment  of  a  person  for  one  department  by  giving  them  a 
joint  appointment. 

When  I  was  chairman  of  the  department  [1969-1974],  I  sat  on 
several  committees  that  were  trying  to  create  a  cancer  center. 
The  notion  was  that  there  would  be  some  new  building,  and  people 
would  be  in  that  building  but  have  their  appointments  in  various 
departments.  I  literally  torpedoed  those  kinds  of  ideas  because 
Biochemistry  would  never  allow  any  of  its  faculty  to  be  in  other 
than  Biochemistry  space. 

Hughes:   As  chairman,  you  could  have  led  a  crusade  to  change  the  policy. 

Berg:    Yes,  but  I  believed  in  it.   Actually,  there  was  one  other  person 
in  Genetics  who  was  really  terrific,  and  that  was  Eric  Shooter. 
Eric  Shooter  came  to  our  department  as  a  postdoc;  he  was  already  a 
pretty  senior  guy.   But  he  came  on  sabbatical  to  work  with 
Baldwin.   And  then  he  went  back  to  England.   Then  Josh  got  a  gift 
from  the  Kennedy  Foundation  to  create  a  neuroscience  program.   He 
recruited  Eric  Shooter  to  come  back  and  head  up  this  neuroscience 
center.   So  Eric  was  given  a  joint  appointment  in  Biochemistry. 

Hughes:   He  was  the  first  in  Biochemistry? 


176 


Berg:    I  can't  remember.   I  think  he  was  the  first  significant  one.   He 
was  located  in  the  genetics  department.   The  genetics  department 
and  Biochemistry  were  separated  by  a  swinging  door.   One  of  our 
graduate  students  elected  to  work  with  Eric  Shooter.   Within  six 
months,  he  asked  to  be  reassigned  to  somebody  in  Biochemistry.   He 
found  being  isolated  uncomfortable.   He  was  mixed  in  with  postdocs 
from  Eric  Shooter's  lab,  and  they  were  doing  genetics.   He  didn't 
feel  part  of  the  genetics  department.   I  think  that  convinced  most 
of  us  that  those  kinds  of  things  don't  work.   If  you  have  a 
faculty  member  in  another  building,  and  he  has  graduate  students, 
those  graduate  students  don't  become  part  of  the  culture  of  the 
main  department.   So  we  resisted  giving  joint  appointments.   In 
fact,  it  was  only  about  two  years  ago  that  we  actually  had  the 
next  such  appointment,  it  was  Gilbert  Lhu,  who  had  been  a  postdoc 
in  my  lab . 

Hughes:   Since  1959?   That's  amazing. 

Berg:    Well,  actually,  the  Shooter  joint  appointment  was  probably  in  the 
mid-sixties . 


Interdisciplinarity 


UCSF 


Hughes:   I'm  thinking  of  the  very  different  model  at  UCSF,  at  least  as  it's 
been  portrayed  to  me,  in  which  departmental  lines  are  quite 
porous.   The  Program  in  Biological  Sciences  [PIBS]  is  totally 
interdisciplinary . 

Berg:    But  you're  talking  about  now.   That  wasn't  the  way  it  was  in  the 
sixties . 

Hughes:  That's  true. 

Berg:  PIBS  was  almost  the  last  five  years. 

Hughes:  Is  it  that  recent? 

Berg:  That's  right.  Mike  Bishop  was  one  of  the  creators  of  PIBS. 

Hughes:   The  idea  of  a  much  less  departmentally  oriented  effort  began  to 
evolve  at  UCSF  in  the  1970s. 

Berg:    I  would  place  it  more  in  the  eighties.   I'll  t.ell  you  why. 


177 

A  Consolidated  Stanford  Graduate  Admissions  Policy  in 
Biology 

Berg:    There  was  a  sense  that  Stanford  biochemistry  as  a  department  was 
beginning  to  compete  for  graduate  students  with  places  that 
offered  more  variety  and  opportunity  for  graduate  students.   While 
we  had  been  extraordinarily  successful  in  recruiting  graduate 
students,  we  were  beginning  to  lose  out  to  places  like  MIT,  UCSF, 
Caltech  that  were  now  creating  larger  entities  into  which  students 
could  enter  and  then  select  where  they  wanted  to  specialize. 
Whereas  we  had  a  very  restricted  set  of  options.   We  had  only  ten 
faculty. 

Hughes:   And  you  did  biochemistry.   [laughter] 

Berg:    That's  right.   And  we  didn't  offer  opportunities  to  somebody  who 
said,  "I'd  like  to  come  to  Biochemistry  but  I  want  to  do  cell 
biology."   So  that  was  one  of  the  impetuses  for  CMGM  [Center  for 
Molecular  and  Genetic  Medicine];  that's  the  whole  thing.   I  tried 
to  get  the  department  to  agree  that  we  would  have  a  schoolwide 
admissions  process,  not  identified  as  biochem.   Until  two  years 
ago,  Biochemistry  said,  "We  don't  want  any  part  of  any  kind  of 
bigger  departmental  thing.   We'll  only  diminish  the  quality  of  the 
students  we  get.   We'll  have  to  compete  with  other  departments. 
Other  departments  will  either  admit  people  that  they  think  are 
good  enough  but  they're  not  our  type." 

So  I  started  a  program  for  admissions  in  CMGM  which  was  to 
run  in  parallel,  because  I  knew  there  was  no  way  that  I  could 
eliminate  the  biochemistry  program  or  its  admissions  process.   So 
I  started  our  own.  We  started  one  under  the  center's  auspices 
which  brought  in  students  and  allowed  them  to  remain 
undif ferentiated,  uncommitted  for  a  year.   They  could  go  to  any 
department  in  the  medical  school. 

Hughes:   Has  that  been  successful? 

Berg:    It  has  now  become  the  program  under  which  our  graduate  students 
are  admitted.   Jim  Spudich,  who  is  the  current  chairman,  finally 
began  to  recognize  that  in  the  program  that  Stanford  had  every 
department  was  going  through  their  own  process,  reviewing 
applications.   It  made  more  sense  to  lump  together  all  the 
"biology"  at  Stanford,  offer  a  program  that  admits  a  student  to 
Stanford  biology,  and  then  give  them  a  year  to  decide  where  to  do 
their  Ph.D.  research.   Some  go  to  biology,  some  go  to 
biochemistry,  some  go  to  pharmacology,  some  go  to  genetics,  and 
that's  much  more  efficient.   Last  year,  we  got  a  terrific  group  of 
students . 


178 


The  thing  that  was  going  on  at  UCSF  was  a  very  strong 
interest.   As  we  began  to  learn  of  the  development  of  the  PIBS 
program,  we  could  see  that  what  was  happening  was  we  were  having 
to  compete  with  people  who  could  offer  students  a  much  richer 
environment. 


Beckman  Center  Programs 


Hughes:   These  interdisciplinary  programs  also  reflect  what's  happening  in 
science.   So  you  have  to  train  people  in  an  interdisciplinary  way. 

Berg:     Yes,  so  at  the  Beckman  Center,  we  retained  the  departmental 

structure,  because  I  think  it's  the  most  efficient  administrative 
unit.   And  probably  for  creating  a  kind  of  social  structure  that 
interacts  and  creates  loyalties,  it's  good.   Overlaying  that,  we 
have  created  a  whole  series  of  interdisciplinary, 
interdepartmental  programs  focused  around  themes. 

We  have  one  that  is  called  Cell  Sciences.   So  it  brings 
people  from  Developmental  Biology,  Biochemistry,  Pharmacology, 
Genetics.   They  all  have  an  interest  in  the  structure  of  a  cell 
and  how  it's  organized,  what  are  the  skeletal  elements,  how  do 
things  get  shunted  around.   We  have  another  one  in  immunology; 
again,  it  brought  together  people  with  very  different  backgrounds, 
including  clinical  departments,  including  people  over  on  the 
campus --biology. 

Now  we  have  four  such  programs .  We  have  one  in  human 
genetics,  one  in  immunology,  one  in  cell  sciences.   We  are  now  in 
the  midst  of  trying  to  create  one  called  structural  biology. 
Because,  just  for  the  reason  you  say,  the  problems  have  become  so 
immense,  that  no  one  individual  can  bring  to  bear  the  kind  of 
insights,  the  technical  expertise,  or  the  knowledge  to  be  able  to 
attack  that  problem  in  a  comprehensive  way.   So  what  you  have  to 
do  is  bring  people  together. 

The  whole  idea  is  to  create  groupings  that  meet  together, 
talk  together,  get  a  grant  together,  have  retreats.   So  I  use 
funding  that  I  get  from  the  Beckman  Foundation  to  seed  these 
programs.   And  sometimes  I  have  to  do  it  by  seduction,  like  the 
cell  sciences  program.   People  were  saying  we  didn't  have  good 
microscopy,  and  cell  biologists  really  need  good  up-to-date 
microscopy.   I  said,  okay,  I  will  create  a  superb  microscopy 
facility,  state  of  the  art,  if  you  guys  will  put  together  a 
program  that  will  build  the  intellectual  activities  around  it.   So 
we  have  a  terrific  facility.  We  expend  $100,000  a  year  to 


179 


maintain  it,  but  people  are  now  doing  advanced  kinds  of  microscopy 
that  they've  never  done  before.  We  have  the  same  thing  for 
molecular  structure.  We  have  twelve  computers  sitting  in  a 
facility  which  is  now  dedicated  to  teaching  people  to  do  molecular 
modeling.   So  you  begin  to  get  people  who  talk  to  each  other. 

So  the  department  lines  now  have  become  what  I  like  to  call 
very  permeable.   People  and  ideas  go  across  departments  easily. 
But  structurally  it's  still  Biochemistry,  still  Genetics,  and  so 
on.   It  took  a  while  to  get  my  colleagues  to  accept  that,  Arthur 
being  one  of  the  most  resistant  to  the-- 


Berg:    --Beckman  Center  concept.   But  I  think  today  he  has  a  very 
different  view  of  it.   I  think  he  sees  it  as  valuable. 


Beckman  Center  for  Molecular  and  Genetic  Medicine 
Origin  of  the  Concept 


Berg:    To  get  to  how  the  CMGM  came  about:  During  the  recombinant  DNA 

controversy,  many  of  us  who  were  participants  were  saying  that  to 
justify  going  ahead  with  something  that  might  be  slightly  risky  we 
need  to  consider  all  the  benefits  and  rewards  that  would  come  from 
it.   And  most  of  those,  we  forecasted,  were  going  to  be  in 
medicine.   Some  predicted  it  would  be  in  agriculture,  but  most 
thought  it  was  medicine  that  was  going  to  be  impacted  the  most. 

I  had  a  visit  from  a  professor  of  medicine  here,  Kenneth 
Melman.   He  said,  "You  go  around  talking  about  all  these  things 
that  are  going  to  happen  in  medicine.   I  don't  have  anybody  in  my 
department  who  even  understands  the  words .   So  if  you  think  that 
we're  going  to  translate  these  great  things  that  you  guys  are 
doing  into  practical  medical  benefits,  you  have  another  guess 
coming.   That  was  a  sobering  thought,  because  I  always  thought  our 
Department  of  Medicine  was  pretty  good.   But  they  were  of  a 
different  vintage.  And  the  field  was  exploding. 

So  Ken  Melman  and  I  began  to  meet  with  the  dean  and  we  said 
what  we  really  need  to  do  is  to  create  a  new  entity  at  Stanford 
which  is  composed  of  people  who  are  trained  in  medicine  but  have 
elected  to  do  science.   Examples  of  such  a  breed  were  Mike  Bishop, 
Harold  Varmus,  Mike  Brown,  Joe  Goldstein.   These  are  people  who, 
following  medical  school,  and  clinical  training,  got  intensive 


180 


basic  science  training  and  elected  to  follow  a  career  in  basic 
science  instead  of  clinical  medicine.  But  they  always  have  in 
mind  the  nature  of  the  biological  problems,  the  medical  problems. 

One  could  foresee  that  if  you  got  these  people  organized  in 
some  way,  you  could  have  very  easy  transfer  of  new  information, 
new  discoveries,  new  technologies,  to  the  "clinic".   In  other 
words,  you  want  to  have  a  group  of  people  who  can  speak  to  both 
physicians  and  the  scientists  and  have  the  respect  of  both;  I 
dubbed  it  the  bench  to  the  bedside  activity. 

We  sold  that  bill  to  the  medical  school  and  the  university. 
The  idea  was  to  create  a  department  of  eight  new  faculty,  all  to 
be  newly  recruited.  We  had  identified  a  group  of  people,  many  of 
whom  came  out  of  M.D.-Ph.D.  programs,  who  were  looking  for  just 
this  kind  of  an  opportunity  to  bridge  across  the  two  fields.   But 
there  was  no  space  to  house  them.   There  was  zero  space.   The 
medical  center  was  filled. 

By  the  time  we  got  around  to  thinking  about  building  a 
building,  we  realized  that  we  had  other  needs  for  space  as  well. 
For  example,  developmental  biology  was  exploding.   So  why 
shouldn't  we  have  a  Department  of  Developmental  Biology?  We  had  a 
Department  of  Physiology  which  had  become  moribund,  literally. 
Physiology  was  being  taught  to  the  medical  students  by  physicians 
who  weren't  at  the  cutting  edge.   And  so  we  argued  that  we  should 
create  a  new  kind  of  department  which  was  dubbed  Molecular  and 
Cellular  Physiology.   The  departments  were  Molecular  and 
Developmental  Biology,  Molecular  and  Cellular  Physiology  and 
Molecular  and  Genetic  Medicine.   Those  names  were  not  accidental 
choices;  they  were  all  chosen  to  drive  home  that  molecular  was  the 
level  we  wanted  to  understand  things  at. 


Raising  Funds 

Berg:    Well,  once  we  decided  to  have  three  departments,  it  was  clear  we 
would  need  big  money  and  a  big  building.   The  Howard  Hughes 
Medical  Institute  was  then  headed  by  Don  Fredrickson,  who  had  been 
the  former  director  of  the  NIH,  and  who  on  his  own  had  been  trying 
to  promote  this  kind  of  training  in  people.   Physicians  who  do 
science.  When  he  heard  that  we  were  going  to  do  that,  Howard 
Hughes  offered  us  $12.5  million  towards  creating  this  kind  of  a 
center.   The  Howard  Hughes  ultimately  added  another  $7.5  million, 
for  a  total  of  $20  million.   They  adopted  the  Department  of 
Molecular  and  Genetic  Medicine  and  called  it  the  Howard  Hughes 
Institute  Unit  of  Molecular  and  Genetic  Medicine.   HHMI  doesn't 


181 


have  departments.   They  also  agreed  to  fund  twelve  investigators, 
plus  the  $20  million  towards  the  construction. 

Actually,  I  got  a  little  of  the  chronology  wrong.   There  was 
a  steering  committee  created  to  try  to  develop  this  concept. 
Besides  me  there  was  the  dean,  Dominick  Purpura,  Stanley  Cohen, 
Hugh  McDevitt,  and  Ken  Melmon,  the  head  of  medicine  at  that  time. 
We  brainstormed  and  focused  on  having  a  center  for  molecular  and 
genetic  medicine. 

At  that  time,  Arnold  Beckman  had  let  it  be  known  that  he 
wanted  to  give  away  his  fortune  before  he  died,  and  that  he  would 
entertain  proposals  for  new  projects.   We  put  together  a  proposal; 
we  invited  him  to  come;  he  spent  three  days;  we  outlined  our 
vision;  he  went  away;  we  never  heard  from  him  again.   We  only 
found  out  later  why. 

In  the  interim,  I  proposed  that  we  have  an  annual  symposium 
called  the  Symposium  on  Molecular  and  Genetic  Medicine.   We 
couldn't  build  a  building  without  money;  we  didn't  have  any 
people,  but  we  ought  to  at  least  start  promoting  the  term,  the 
concept,  on  a  national  scale.   So  we  had  several  terrific  national 
symposia  on  this  theme.   Fredrickson  came  to  one  of  them,  and  we 
told  him  our  plans.   He  was  going  to  become  the  president  of  the 
Howard  Hughes  Medical  Institute  and  he  would  recommend  investing 
$12.5  million  in  the  CMGM  and  ultimately  more.   So  we  had  a  start 
on  the  funding.   But  no  word  from  Beckman  at  all. 

We  had  $12.5  million;  the  building  was  going  to  cost 
something  like  $40  million,  and  because  we  were  so  far  short  of 
the  cost  the  trustees  would  not  allow  us  to  move  ahead,  either  to 
plan  or  do  anything.  But  Beckman  remained  silent.   Only  later  did 
we  learn  that  he  was  not  about  to  fund  a  project  under  the  aegis 
of  a  "directorate",  our  steering  committee.   He  felt,  "That's  not 
the  kind  of  organization  I  want  to  support.   I  want  to  see  one 
person  who  has  committed  his  energy,  vision  and  activity 
full-time,  and  whom  I  believe  and  trust.   I  won't  give  money  to 
something  that  is  going  to  be  run  by  a  group."  With  no  word  from 
Beckman,  the  project  was  moribund.  At  that  point  Don  Kennedy  came 
to  me  and  he  said,  "Either  you  become  the  director,  or  we  drop  the 
whole  thing." 

I  had  been  asked  several  times  to  be  the  director.   I  had 
refused;  I  didn't  want  to  do  that.   I  was  perfectly  willing  to 
spend  time  on  this  committee  developing  the  concept.   But  I  did 
not  want  to  be  the  lead.   But  when  Kennedy  threatened  to  trash  the 
whole  project,  I  agreed. 


182 

Within  weeks,  I  called  Arnold  Beckman  and  I  said,  "There  has 
been  a  change  in  our  plans.   I  have  become  the  director.   I'd  like 
to  come  to  talk  to  you  about  my  vision  for  what  will  happen.   And, 
he  invited  me  to  come.   I  asked  Don  and  Arthur  to  join  me.   We  met 
with  Arnold  Beckman  for  twenty  minutes.  And  two  weeks  later  he 
called  and  said  he'd  give  us  the  twelve  and  a  half  million  dollars 
we  asked  him  for.   Later,  when  the  big  announcement  about  his  gift 
was  made,  he  was  asked  why  he  was  making  this  gift  considering 
that  he  had  no  affiliation  with  Stanford.   He  said  he  knew  me;  he 
knew  Arthur;  and  that  he  had  complete  confidence  in  us.   If  I  was 
willing  to  take  the  responsibility  of  directing  this  whole  effort, 
then  that  was  good  enough  for  him. 

Hughes:   Quite  a  compliment. 

Berg:    It  was.   So  now  we  had  thirty  some  odd  million  dollars,  and  then 
Arnold  Beckman  helped  us  raise  some  money  from  other  sources, 
which  he  had  access  to.   He  persuaded  SmithKline  Beecham,  then 
SmithKline  Beckman,  to  give  us  $8  million  dollars.   It's  rare  that 
an  industrial  company  gives  you  money  to  build  a  building.   So  we 
got  the  Center  built. 


Berg's  Strategic  Decisions 

Berg:    There  were  several  strategic  decisions  which  I  had  to  make,  which 
in  retrospect  I  believe  were  the  right  ones.   I  should  have  said 
before:  When  I  was  asked  to  become  director,  I  said  I  had  two 
conditions.   One  was  that  the  school  would  allow  the  building 
planning  and  construction  to  go  ahead  even  if  we  didn't  have  all 
the  money  in  hand.   Kennedy  agreed  to  that.  And  second, 
biochemistry  had  to  become  part  of  the  center.   I  said  there  was 
no  way  I  was  going  to  devote  the  next  five  or  ten  years  of  my  life 
and  leave  my  colleagues  sitting  in  the  old  building  in  antiquated 
space.   Well,  it  wasn't  quite  antiquated,  but  they  had  to  be  part 
of  the  center.   I  also  believed  that  there  would  be  an  enormous 
drawing  card  for  recognition  of  the  center  if  the  famous 
biochemistry  department  was  going  to  be  in  it.   That  meant  the 
building  had  to  be  bigger  and  cost  more  money.   The  cost  was  going 
to  be  up  around  $50  to  $60  million.   So  the  job  of  fund  raising 
came  to  be  a  big  job.  But  we  raised  almost  $96  million  in  those 
three  years.   I  spent  a  lot  of  time  on  the  road,  meeting  with 
people,  persuading  them.   Dave  Kern  was  very  important  and  Arnold 
helped  as  well.   But,  eventually  we  got  the  money  to  go  ahead. 

Another  strategic  decision  was  the  three  new  departments  in 
that  building,  besides  Biochemistry,  were  to  be  comprised  of 


183 


people  who  were  recruited  from  outside,  not  who  were  already  ar 
Stanford  in  existing  departments.   There  were  two  reasons  for  this 
decision:  you  could  gather  up  all  the  stars  that  existed  in 
Stanford  departments  and  move  them  to  the  Center  and  leave  behind 
a  wasteland.   That  was  the  worst  thing  you  could  have  done  because 
it  would  have  really  created  a  lot  of  anger  and  anxieties.   Those 
ransacked  departments  would  now  have  had  to  go  out  and  recruit 
replacements. 

We  had  the  advantage  in  being  able  to  do  that.   We  had  a  new 
building;  we  had  a  terrific  concept.   So  I  said  we  were  not  going 
to  move  any  people.   And  Stan  Cohen  became  my  bitter  enemy,  in 
part  because  he  was  on  that  steering  committee.   I  recall  once 
when  I  came  back  from  a  trip  to  find  that  Stan  had  carved  out 
space  for  himself  in  that  building,  as  had  Melmon  and  a  few  other 
people.   When  I  came  back  I  raised  hell.  When  I  became  director, 
I  said,  "There  are  not  going  to  be  any  moves."  Obviously,  I  was 
vulnerable  because  I  had  insisted  that  Biochemistry  would  move  to 
the  Center. 

Hughes:   How  did  you  handle  that? 

Berg:    I  said  straight  off  that  that  was  my  price  for  being  Director.   I 
had  made  it  clear  that  I  was  not  going  to  do  it  without 
benefitting  my  colleagues  who  I  was  convinced  would  enhance  the 
standing  of  the  Center  and  thereby  aid  in  the  recruiting  of  new 
people.   I  think  that  was  correct,  indeed  Arnold  Beckman  agreed. 

Hughes:   The  other  departments  were  created  de  novo? 

Berg:    Yes.   So  we  set  about  recruiting.   Eventually,  we  had  to  give  in  a 
little  on  the  decision  of  nobody  from  Stanford.   The  building  was 
going  to  take  two  and  a  half  years  to  build.   Howard  Hughes  had 
committed  money  and  positions,  and  they  kept  saying  to  us,  "Who 
are  the  Howard  Hughes  investigators?"  Well,  you  couldn't  recruit 
anybody  to  a  building  that  was  not  going  to  be  available  for  two 
and  a  half  years.   So  we  eventually  identified  a  few  people  within 
the  school  who  almost  surely  would  have  been  Howard  Hughes 
investigators  anywhere  else.   The  three  of  them  were  Jerry 
Crabtree,  Irving  Weissman  and  Gary  Schodnick.   That's  all  we  did. 
They  became  the  founders  of  the  Howard  Hughes  unit,  which 
ultimately  reached  twelve  people.   So  everyone  else  was  newly 
recruited- -new  chairmen,  and  new  faculty. 

To  this  day,  I'm  absolutely  convinced  my  decision  was  right 
because  had  we  filled  the  Center  with  the  people  we  had,  we  never 
would  have  been  able  to  create  the  kinds  of  connections  back  to 
the  departments.   The  whole  idea  was  to  use  the  Beckman  Center  as 
the  focus  for  this  concept  of  molecular  genetic  medicine  and  build 


184 


bridges  to  existing  people.  So  we  recruited  people  who  could 
relate  to  or  connect  to  the  strength  we  already  had,  and  that 
happened. 

The  second  important  decision  came  from  the  recognition  that 
there  was  a  political  problem.   There  were  a  lot  of  people  who 
wanted  to  be  in  the  Beckman  Center,  new  space.   We  frequently 
heard,  "I  do  molecular  genetic  medicine;  why  shouldn't  I  be  in 
that  building?"  We  had  to  admit,  "We  all  do  molecular  genetic 
medicine. " 


The  Program  in  Molecular  and  Genetic  Medicine 

Berg:     It  turns  out  there  are  a  hundred  and  ninety  people  who  do  what 

could  be  called  molecular  and  genetic  medicine.   We  can't  all  be 
in  one  building.   Instead,  we  can  be  part  of  and  intellectual 
group  called  the  Program  in  Molecular  and  Genetic  Medicine  [PMGM] . 
Members  of  PMGM  differ  one  from  another  in  where  we  live.   In 
terms  of  funding,  access  to  facilities,  teaching  opportunities, 
you  name  it,  everybody  has  equal  access,  and  that's  what  we  have. 
In  a  sense  the  Program  in  Molecular  and  Genetic  Medicine  serves  as 
an  umbrella  for  those  interested  in  molecular  and  genetic 
approaches  to  biological  questions. 

The  physical  focus  is  the  Center.   The  Program  encompasses 
people  in  the  basic  science  departments,  the  clinical  departments, 
chemistry,  biology,  applied  physics.   There  are  now  196  people  who 
declare  themselves  as  members  of  the  program.   They  are  invited  to 
all  kinds  of  PMGM  functions.   They've  organized  these 
interdisciplinary  units  on  various  themes.   They  have  their  own 
retreats  seminar  programs.   We  try  constantly  to  meld  basic 
science  with  clinical  activities.  We  have  workshops  where  we  have 
brought  physicians  in  to  tell  us  about  bone  marrow  transplants;  we 
have  immunologists  and  gene  therapists  who  want  to  use  bone  marrow 
transplants. 

It  has  been  an  exciting  enterprise.   When  I  retired  from  my 
professorship  in  1998  the  dean  said,  "I  want  you  to  remain  as  the 
director  of  Beckman."  Actually,  I  don't  direct  anything  other 
than  to  try  to  create  new  kinds  of  interactions.   One  has  to  be 
clever  and  imaginative  to  get  people  to  come  together  when  they're 
all  busy  doing  their  own  thing. 


185 


Stanford  Biochemistry's  Asilomar  Conferences 


Berg:    The  Asilomar  conference  was  my  invention  when  I  became  chairman  of 
the  biochemistry  department.   We  needed  a  way  to  get  people  in  the 
department  to  talk  to  each  other  about  their  research.   We  used  to 
do  it  in  the  department  library  but  people  were  sneaking  out  to 
the  lab  or  going  home  early.   It  was  clear  that  the  only  way  it 
would  work  was  to  go  away  from  Stanford  and  its  distractions.   We 
started  the  Asilomar  conferences  in  1970  or  '71,  and  they've  been 
running  ever  since,  not  always  at  Asilomar.   Now  it's  the 
highlight  of  the  scientific  year. 

Hughes:   Who  goes? 

Berg:     Everybody;  there  were  175  people  that  went  to  the  Stanford  Center 
at  Fallen  Leaf  Lake  at  this  last  one.   I  just  got  back  last  week. 
Every  group  is  given  a  slot  of  time,  usually  60-90  minutes,  for 
presentations.   The  head  of  the  research  group  programs  what  the 
people  in  his  group  will  present.   There  is  a  lot  of  time  for 
discussion.   Those  that  don't  get  a  chance  to  talk  about  their 
work,  do  it  through  poster  sessions.   Most  people  feel  it's  the 
best  scientific  meeting  they've  attended  in  the  year,  because 
there  is  terrific  research  going  on.   It  lasts  three  days,  and  is 
intensive,  nose-to-nose.   People  eat  every  meal  together.   That 
format  has  been  copied  all  over  the  country.   UCSF  has  started  it; 
Berkeley  has  started  it;  Harvard,  MIT.   They  all  have  retreats; 
they  all  have  found  local  places  where  you  can  go  and  hide. 

Hughes:   Stanford's  was  the  first? 
Berg:    We  were  the  first. 


Greatest  Contribution 


Hughes:   I  have  one  more  question,  but  is  there  anything  you  want  to  say 
before  that? 


Berg:    No. 

Hughes:   What  do  you  consider  to  be  your  greatest  contribution? 

Berg:    Some  years  ago  there  was  a  celebration  of  Severe  Ochoa's 

seventieth  birthday,  in  Spain,  and  all  of  his  former  students, 
colleagues  and  friends  were  invited  to  go.   I  think  Arthur  was  one 
of  the  co-organizers.   I  was  not  a  student  of  Ochoa;  I  had  never 


186 


had  any  collaboration  with  Ochoa.   But  for  some  reason  early  on  he 
took  a  liking  to  me,  and  he  always  treated  me  like  one  of  his 
"children",  so  I  was  invited  to  go  to  Spain  for  this  symposium. 

We  were  each  asked  to  write  an  essay.   Most  people  just 
wrote  a  paper  about  some  of  their  recent  work.   I  tried  to  do 
something  different.   I  tried  to  ask  what  is  the  measure  of 
somebody's  greatness  or  the  contribution  they  made.   What  meant 
the  most  to  me  is  the  long-term  impact  of  ones  work  or  efforts. 
Pebbles  dropped  in  water  produce  a  succession  of  ripples,  and  one 
might  ask  what  those  ripples  created.   Is  it  just  the  impact  of 
the  stone,  or  is  it  the  effect  of  the  ripples  that's  important?   I 
tried  to  think  of  it  in  terms  of  who  have  these  people  trained? 
Who  are  the  people  who  have  brought  their  style,  their  message, 
their  philosophy,  their  whole  concept  of  science  and  taught  their 
students  that  way. 

I  had  the  idea  of  trying  to  measure  this  by  way  of  a  star 
chart.   An  individual  would  be  represented  as  a  dot.   The  people 
that  person  trained  would  be  represented  by  radiating  lines  from 
that  dot.   The  length  of  the  line  was  to  be  proportional  to  the 
impact  or  success  that  person  had  achieved;  that  would  be  a 
measure  of  his  or  her  accomplishment.   Then  those  people  would 
have  trained  somebody,  and  there  would  have  been  branches  off  that 
line.   You  would  have  created  a  halo  around  this  dot.   The  density 
of  the  halo  would  be  a  measure  of  what  the  person  had  contributed 
to  science,  and  how  far  the  halo  came  would  be  some  measure  of  how 
successful  his  progeny  were  in  science. 

I  tried  to  do  that  kind  of  representation.   Ochoa  was  one, 
and  I  was  going  to  have  about  five  or  six  other  heroes  in  science 
to  get  a  measure  of  their  impact.   Some  of  them  have  done  great 
science  and  got  the  Nobel  Prize  but  hardly  ever  trained  anybody, 
or  ended  up  with  flunkies  who  never  went  on  to  do  anything  else. 
I  wanted  to  be  able  to  illustrate  that,  but  Arthur  talked  me  out 
of  doing  it.   He  said,  "You  will  make  enemies  that  will  last  a 
long  time.   How  are  you  going  to  be  able  to  identify  who  this  line 
is,  and  who  the  short  stub  is?"  I  thought  better  of  it,  and  I 
didn't  do  it.   But  the  concept  has  always  stuck  in  my  mind. 

When  you  ask  me  what  has  been  my  greatest  contribution,  I 
think  it's  the  students  that  have  come  out  my  lab,  and  the 
tremendous  science  that  many  of  them  have  done,  and  the  affection 
ad  relationship  that  exist  between  us.   I'm  still  in  touch  with  so 
many  of  these  students.   They  put  on  a  party  for  me  on  my  sixty- 
fifth  birthday,  which  would  blow  you  away.   The  party  took  over 
the  Mark  Hopkins  Hotel,  and  the  DeYoung  Museum.   The  museum  has  a 
magnificent  hall  hung  with  tapestries,  which  served  for  the 
dinner.   The  organizers,  all  students,  raised  $160,000  for  this 


187 


party.   Former  students  and  postdocs  came  from  Europe,  Asia,  all 
over  the  world.   It  was  an  incredible  party.  Anyway,  that 
relationship  and  admiring  what  they've  accomplished  to  me  is  the 
most  important  thing. 

Many  of  them  say  that  the  way  we  did  science  in  the  lab  at 
Stanford  has  always  influenced  the  way  they  do  science  and  what 
they  try  to  convey  to  their  students.   To  me,  that  is  probably 
more  than  just  the  few  things  that  I've  done,  or  done  with  them, 
because  that  radiating  effect  is  going  to  influence  much  more  of 
science  than  I  could  have  done  alone. 

I  think  about  our  department  in  the  same  way.   Our 
department  has  done  a  lot  of  great  science;  It's  not  possible  to 
talk  about  contemporary  science  without  running  into  someone  who 
was  a  postdoc  or  student  at  Stanford.   That's  a  very  satisfying 
feeling. 


Hughes:   Thank  you. 


Transcribed  by  Quandra  McGrue 
Final  Typed  by  Grace  Robinson 


188 
TAPE  GUIDE--Paul  Berg 


Interview  1:  July  15,  1997 

Tape  1,  Side  A  1 

Tape  1,  Side  B  11 

Tape  2,  Side  A  22 

Tape  2,  Side  B  31 

Tape  3,  Side  A  40 
Tape  3,  Side  B  not  recorded 

Interview  2:  August  12,  1997 

Tape  4,  Side  A  42 

Tape  4,  Side  B  51 

Tape  5,  Side  A  61 

Tape  5,  Side  B  71 
Tape  6,  Side  A  not  noted  (only  5  minutes  recorded) 
Tape  6,  Side  B  not  recorded 

Interview  3:  September  30,  1997 

Tape  7,  Side  A  80 

Tape  7,  Side  B  90 

Tape  8,  Side  A  100 

Tape  8,  Side  B  110 

Tape  9,  Side  A  120 
Tape  9,  Side  B  not  recorded 

Interview  4:  November  5,  1997 

Tape  10,  Side  A  128 

Tape  10,  Side  B  138 

Tape  11,  Side  A  147 

Tape  11,  Side  B  158 

Tape  12,  Side  A  168 

Tape  12,  Side  B  179 


SAMPLE  EDITED  PAGE 
189 

(from  transcript,  with 

narrator's  editing) 
141 


molecule,  and,  inside  the  cell,  it  jjuutfjlHLLwLy  circularizes  and  the 

TV 
nicks  mi  muuuuh.  between  the  ends  get  closed.   And  new  it  functions 


as  a  circular  DNA  molecule. 


Creating  Artificial  Cohesive  Ends 


Berg:    So,  the  concept  of  sticky  ends  wpe  already  tteste .   If  you  want  to 
join  two  different  molecules  together,  it  doesn't  take 


genius  to  figure  out  that  if  you  can  &e*w«41y  create  artificial 

frlA  \~Tr4t  t+ I*  j 
ends  that  are  complementary  to  each  other  Cftttt  the  two  will  come 

A 
together.   Right?   No  big  deal. 


So,  th*  q\igf7ti"n  in.  if  ynn  ha"?  a  paggp  vf  TW  ft  nrH  yn 


have.  OV40  DMA  D,  if  you  put  tails  of  A1  on  ••e' and  tails  of  T  on 

j^  f\ 


-e  J 
••e' 


rhni'.nfthrr,.  and  jpi:  mix  them,  the  A's  and  T's  will  form  double 

J*W  J>  *-***> 
helices,  and  the  two  molecules  will  come  together.   Tw^s  cannot 


<?f 
join  to   itself ^TJssPcould  have  used  G's   and  C's,   but  A's   and  T's 

were   easier   to   add. 


We  already  knew  how  to  add  tails  onto  tifewEe  DNA  molecules 

because  there  is  an  enzyme  that  had  been  described  which  is 

,.*- 

present  in  flfc  calf  thymus  '  a«*d  has  an  interesting  physiological 


rv 

f  unction,  but  ftR;  was^  not  known  at  «wt  time.   It  w*6  DNA 

t'    > 
polymerase,  but  "K*6  a  dumb  DNA  polymerase.   It  doesn't  need  a 

5      Tr, 

template.   If  you  give  it  any  one  of  the  four  deoxynucleo^idef/^  it 

"p<  K  *  *)*  j-k  '±4. 
will  add  Jt£  on  to  the  end  of  the  DNA  molecule  jfd  JU-UICESSEB.   So,  if 


.     - 
r  DNA  molecule  A,  and--jfi>i  put  in  deoxyATP  and  this 

O/ 


*^S.  POy  -v»f  i   ,  ** 

enzyme,    it  wiil  pfel^naerrpc  A's  onto  the  two  3/prime  ends   of  this 

A 

DNA.  And  if  you  do  it  with  deoxyTTP 


in  ill:  "lull    T's  -oar.     By  regulating  the  time 


the  reaction,  you 


APPENDIX 


A  Paul  Berg,  CV  and  Publications  190 

B   Selections  from  Berg  papers,  courtesy  Green  Library, 

Stanford  University  208 

C   "The  1980  Nobel  Prize  in  Chemistry,"  Science,  vol.  210,  21 

November  1980  241 


190  APPENDIX  A 


PAUL  BERG 

Professor  of  Biochemistry 

Stanford  University  School  of  Medicine 

Stanford,  California  94305 

Bom:  June  30,  1926,  New  York,  New  York 

Address:          838  Santa  Fe,  Stanford.  CA  94305 
Soc.  Sec.  #:     095-18-1653 

Education: 

1948  B.S.,  Pennsylvania  State  University 

1952  Ph.D.,  Western  Reserve  University 

Professional  Background: 

1950-52  Predoctoral  Research  Fellow,  National  Institutes  of  Health. 

1952-54  Postdoctoral  Research  Fellow,  American  Cancer  Society,  Dr.  H.  M. 

Kalckar,  Institute  of  Cytophysiology,  Copenhagen,  Denmark  and  Dr. 

Arthur  Kornberg,  Washington  University  School  of  Medicine,  St.  Louis,  MO 
1954  Scholar  in  Cancer  Research,  American  Cancer  Society,  Department  of 

Microbiology,  Washington  University  School  of  Medicine. 
1 955-59  Assistant  to  Associate  Professor  of  Microbiology,  Washington  University 

School  of  Medicine 
1959-60  Associate  Professor  of  Biochemistry,  Stanford  University  School  of 

Medicine 
1960  Professor,  Department  of  Biochemistry,  Stanford  University  School  of 

Medicine. 
1969-74  Chairman,  Department  of  Biochemistry,  Stanford  University  School  of 

Medicine 

1970-94  Sam,  Lulu  and  Jack  Willson  Professor  of  Biochemistry. 

1973-83  Non-Resident  Fellow  of  Salk  Institute. 

1985  Director,  Beckman  Center  for  Molecular  and  Genetic  Medicine. 

1994  Named  Vivian  K.  and  Robert  W.  Cahill  Professor  in  Cancer  Research 

Honors: 

Eli  Lilly  Award  in  Biochemistry  (1959) 

California  Scientist  of  the  Year  (1963) 

National  Academy  of  Sciences  (1966) 

American  Academy  of  Arts  and  Sciences  (1966) 

Henry  J.  Kaiser  Award  for  Excellence  in  Teaching  at  Stanford  University  School  of 

Medicine  (1969,  1972) 

Distinguished  Alumnus  Award,  Pennsylvania  State  University 
V.D.  Mattia  Prize  of  the  Roche  Institute  for  Molecular  Biology 
Institute  of  Medicine,  National  Academy  of  Science  (1974) 
President,  American  Society  of  Biological  Chemists  (1975) 
Honorary  Doctor  of  Science,  University  of  Rochester  and  Yale  (1978) 
Sarasota  Medical  Awards  for  Achievement  and  Excellence  (1979) 
Annual  Award  of  the  Gairdner  Foundation  (1980) 
Nobel  Award  in  Chemistry  (1980) 
Albert  Lasker  Basic  Medical  Research  Award  (1980) 
New  York  Academy  of  Sciences  Award  (1980) 
Foreign  Member,  French  Academy  of  Sciences  (1981) 
American  Association  for  the  Advancement  of  Science  Scientific  Freedom  and 

Responsibility  Award  (1982) 


191 


National  Medal  of  Science  (1983) 

American  Philosophical  Society  (1983) 

Associate  Member  of  EMBO  (1984) 

Honorary  Doctor  of  Science,  Washington  University,  St.  Louis  (1986) 

National  Library  of  Medicine  Medal  (1986) 

American  Academy  of  Achievement  (1988) 

Honorary  Doctor  of  Science,  Oregon  State  University  (1989) 

Special  Achievement  Award,  Odyssey  Biomedical  Corporation 

Fellow  of  American  Association  for  the  Advancement  of  Science  (1991) 

Honorary  Member  of  the  Academy  of  Natural  Sciences  of  the  Russian  Federal  Republic 

(1991) 

Foreign  Member  of  the  Royal  Society  (1992) 
Fellow,  American  Academy  of  Microbiology  (1992) 
Honorary  Member  Alpha  Omega  Alpha  Honor  Medical  Society  (1992) 
Honorary  Member  AMBO/AMBL  (1994) 

Honorary  Doctor  of  Science,  Pennsylvania  State  University  (1995). 
Member,  Pontifical  Academy  of  Sciences  (1996) 

Special  Appointments: 

Editor,  Biochemical  and  Biophysical  Research  Communications 

Member,  NIH  Study  Section  on  Physiological  Chemistry 

Member,  Journal  of  Molecular  Biology  Editorial  Board 

Member,  Board  of  Scientific  Advisors  of  Jane  Coffin  Childs  Foundation  for  Medical 

Research 
Member,  Advisory  Boards  to  National  Institutes  of  Health,  American  Cancer  Society, 

National  Science  Foundation,  Massachusetts  Institute  of  Technology,  and  Harvard 

University 
Elected  to  the  Council  of  National  Academy  of  Science  and  to  the  Scientific  Advisory 

Board  of  the  Welch  Foundation 
Member  and  Chairman,  International  Advisory  Board  of  the  Basel  Institute  of 

Immunology 

Chairman,  Whitehead  Institute  Board  of  Advisory  Scientists 
Chairman,  National  Advisory  Committee,  Human  Genome  Project 
Trustee,  Rockefeller  University 

Chairman,  Board  of  Directors,  National  Foundation  for  Biomedical  Research 
Chairman,  Public  Policy  Committee,  American  Society  for  Cell  Biology 
Advisory  Editorial  Board,  Molecular  Medicine  Today 
Advisory  Panel,  Human  Genome  Education  Program 
Whitehead  Institute  Board  of  Associates 
Scientific  Advisory  Committee,  Research! America 
International  Scientific  Advisory  Board  (ISAB) 
Chairman,  Scientific  Advisory  Board,  Beckman  Foundation 
Advisory  Board,  McGovem  Institute  for  Brain  Research 

Commercial  and  Civic  Activities 

Founder  and  Principal  Scientific  Advisor,  Schering-Plough's  DNAX  Research  Institute 

Director,  Affymetrix 

Consultant,  Bay  Area  Bioscience  Center 

Consultant,  Santa  Clara  County  Biotechnology  Education  Partnership 

Council  of  Advisors  to  San  Francisco  Unified  School  District 

Advisory  Board,  ARISE  (American  Renaissance  in  Science  Education) 


54 


192 

BIBLIOGRAPHY 
Paul  Berg 


1.  Berg,  P.  and  Joklik.  W.K.  Enzymatic  phosphorylation  of  nucleose  diphosphates.  J. 

Biol.  Chem.  210:  657. 

1955 

2.  Berg,  P.,  Participation  of  adenyl-acetate-activating  system.  J.  Am.  Chem.  Soc.  77: 

3163. 

1956 

3.  Berg,  P.  and  Newton,  G.  Adenyl-acetates  in  the  activation  of  acvl  groups.  Fed.  Proc. 

15:713. 

4.  Berg,  P.  Acyl  adenylates:  The  synthesis  and  properties  of  adenyl  acetate.  J.  Biol. 

Chem.  222:  1015. 

5.  Berg,  P.  Acyl  adenylates:  The  interaction  of  adenosine  triphosphate  and  L- 

methionme.  J.  Biol.  Chem.  222:  1025. 

6.  Berg,  P.,  Acyl  adenylates:  An  enzymatic  mechanism  of  acetate  activation.  J.  Biol. 

Chem.  222:991. 

1957 

7.  Bers,  P.  Chemical  synthesis  and  enzymatic  utilization  of  adenyl  amino  acids.  Fed 

Proc.  16:  658. 

8.  Berg,  P.,  Nismann,  B.,  Bergmann,  F.H.  Observations  on  amino  acid-dependent 

exchanges  of  inorganic  pyrophosphate  and  ATP.  B.B.A.  26:  639. 

1958 

9.  Ofengand,  J.,  Bergmann,  F.,  Berg,  P.  Enzymatic  synthesis  of  an  RNA-amino  acid 

complex.  Fed.  Proc.  17:  1123. 

10.  Berg,  P.,  and  Ofengand,  E.J.  An  enzymatic  mechanism  for  linking  amino  acids  to 

RNA.  PNAS44:78. 

1 1 .  Berg,  P.  Studies  on  the  enzymatic  utilization  of  amino  acyl  adenylates:  The 

formation  of  adenosine  triphosphate.  J.  Biol.  Chem.  233:  601. 

12.  Berg,  P.  The  chemical  synthesis  of  amino  acyl  adenylates.  J.  Biol.  Chem.  233:  608. 

13.  Berg,  P.  Role  of  magnesium  in  acetyl  coenzyme  A  formation  by  acetothiokinase. 

Science  129:  3353. 

1959 


193 

14.  Preiss,  J.,  Berg.  P.,  Ofengand,  E.J.,  Bergmann,  F.H.,  and  Dieckmann,  M.  The 

chemical  nature  of  the  RNA-amino  acid  compound  formed  by  amino  acid- 
activating  enzymes.  PNAS  45:  319. 

15.  Bergmann,  F.H.,  Berg,  P.,  Preiss,  J.,  Ofengand,  E.J.,  and  Dieckmann,  M.  Enzymatic 

activation  and  transfer  of  amino  acids  to  PJs'A.  Fed.  Proc.  18:  75 1 . 

1960 

16.  Preiss,  J.,  and  Berg,  P.  Incorporation  of  ATP-C14  into  polyribonucleotides.  Fed. 

Proc.  19:  317. 

1961 

17.  Lagerkvist,  U.,  Berg,  P.,  Dieckmann,  M.  and  Platt,  F.W.  Terminal  nucleotide 

sequences  in  ammo  acid-acceptor  RNA.  Fed.  Proc.  20,  No.  I. 

18.  Berg,  Paul,  Bergmann,  Fred  H.,  Ofengand,  E.J.  and  Dieckmann,  M.  The  enzymic 

synthesis  of  amino  acyl  derivatives  of  ribonucleic  acid.  I.  The  mechanism  of 
leucyl-,  valyl-,  isoleucyl-,  and  methionyl  ribonucleic  acid  formation.  J.  Biol. 
Chefn.  236:  1726. 

19.  Bergmann,  Fred  H.,  Berg,  Paul  and  Dieckmann,  M.  The  enzymic  synthesis  of  ammo 

acyl  derivatives  of  ribonucleic  acid.  II.  The  preparation  of  leucyl-,  valyl-, 
isoleucyl-,  and  methionyl  ribonucleic  acid  synthetases  for  Escherichia  coli.  J. 
Biol.  Chem.  236:  1735. 

20.  Ofengand,  E.J.,  Dieckmann,  M.  and  Berg,  P.  The  enzymic  synthesis  of  amino  acyl 

derivatives  of  ribonucleic  acid.  III.  Isolation  of  amino  acid-acceptor  ribonucleic 
acids  from  Escherichia  coli.  J.  Biol.  Chem.  236:  1741. 

21.  Preiss,  J.,  Dieckmann,  M.  and  Berg,  Paul.  The  enzymic  synthesis  of  amino  acyl 

derivatives  of  ribonucleic  acid.  IV.  The  formation  of  the  3'-hydroxyl  terminal 
trinucleotide  sequence  of  amino  acid-acceptor  ribonucleic  acid.  J.  Biol.  Chem. 
236:  1748. 

22.  Berg,  P.  Specificity  in  protein  synthesis.  In  Annual  Review  of  Biochemistry 

(J.M.Luck,  F.W.Allen  and  G.'MacKinney,  eds.).  Vol..  30,  Annual  Reviews,  Inc., 
Palo  Alto,  CA,  pp.  293-324. 

62 


23.  Chamberlin,  Michael  and  Berg,  Paul.  Deoxyribonucleic  acid-directed  synthesis  of 

ribonucleic  acid  by  an  enzyme  from  Escherichia  coli..  PNAS  48:  81. 

24.  Wood,  William  B.  and  Berg,  Paul.  The  effect  of  enzymatically  synthesized 

ribonucleic  acid  on  amino  acid  incorporation  by  a  soluble  protein-ribosome  system 
from  Escherichia  coli..  PNAS  48:  94. 

25.  Berg,  Paul.  Assay  and  preparation  of  yeast  aceto-CoA-kinase.  In  Methods  in 

Enzymology  (S.P.  Colowick  and  N.O.  Kaplan,  eds.).  Vol.  V,  Academic  Press  Inc., 
New  York,  pp.  461-466. 


194 

26.  Wood,  W.B.  The  effect  of  enzymatically  synthesized  RNA  on  ammo  acid 

incorporation  by  a  soluble  protein-ribosome  system  from  E.  coli.  Fed.  Proc.  21, 
No.  2. 

27.  Chamberlin,  M.  and  Berg,  P.  Enzymatic  synthesis  of  specific  polyribonudeotides. 

Fed.  Proc.  21,  No.  2. 

28.  Berg,  P.,  and  Lagerkvist,  U.  An  attempt  to  correlate  amino  acid  specificity  with 

terminal  nucleotide  sequence  in  amino  acyl  RNA  formation.  In  Acides 
Ribonucleiques  et  Polyphosphates,  Structure,  Synthese  et  Fonctions.  Colloq. 
Intern.  Centre  Nat.  Rech.  Sci.  No.  106,  Strasbourg,  Editions  du  Centre  National  de 
la  Recherche  Scientifique,  Paris,  pp.  259-276. 

29.  Berg,  Paul,  Lagerkvist,  U.  and  Dieckmann,  M.  The  enzymic  synthesis  of  amino  acyl 

derivatives  of  ribonucleic  acid.  V.  Nucleotide  sequences  adjacent  to  the  .  . 
pCpCpA  end  groups.  J.  Mol.  Biol.  5:  139. 

30.  Berg,  P.,  Lagerkvist,  Ulf  and  Dieckmann,  M.  The  enzymic  synthesis  of  amino  acyl 

derivatives  of  ribonucleic  acid.  VI.  Nucleotide  sequences  adjacent  to  the  .  . 
pCpCpA  end  groups  of  isoleucine-  and  leucine-specific  chains.  J.  Mol.  Biol.  5: 
159. 

3 1 .  Berg,  Paul.  Studies  on  the  contribution  of  nucleic  acids  to  the  specificity  of  protein 

synthesis.  In  Basic  Problems  in  Neoplastic  Disease  (A.  Gallhom  and  E. 
Hirschberg,  eds.).  Columbia  University  Press,  New  York,  pp.  15-34. 

1963 

32.  Berg,  P.,  Fancher,  H.  and  Chamberlin,  M.  The  synthesis  of  mixed  polynucleotides 

containing  ribo-  and  deoxyribo-nucleotides  by  purified  preparations  of  DNA 
polymerase  from  Escherichia  coli.    In  Informational  Macromolecules  (H.J.  Vogel. 
V.  Bryson  and  J.O.  Lampen,  eds.).  Academic  Press  Inc.,  New  York,  pp.  467-483. 

33.  Littauer,  U.Z.,  Muench,  K.,  Berg,  P.,  Gilbert,  W.  and  Spahr,  P.P.  Studies  on 

methylated  bases  in  transfer  RNA.  Cold  Sprins  Harbor  Symposia  on  Quantitative 
Biology  XXVIII:  157-159.. 

34.  Chamberlin,  Michael.  Baldwin,  Robert  L.  and  Berg,  Paul.  An  enzymically 

synthesized  RNA  of  alternating  base  sequence:  physical  and  chemical 
characterization.  J.  Mol.  Biol.  7:  334. 

35.  Chamberlin,  M.  and  Berg,  P.  Studies  on  DNA-directed  RNA  polymerase:  formation 

of  DNA-RNA  complexes  with  single-stranded  6X  174  DNA  as  template.  Cold 
Spring  Harbor  Symp.  Quant.  Biol.  28:  67. 

36.  Wood,  W.B.  and  Berg,  P.  Studies  on  the  "messenger"  activity  of  RNA  synthesized 

with  RNA  polymerase.  Cold  Spring  Harbor  Symp.  Quant.  Biol.  28:  237. 

1964 

37.  Chamberlin,  M.,  and  Berg,  P.  Mechanism  of  RNA  polymerase  action:  Formation  of 

DNA-RNA  hybrids  with  single-stranded  templates.  J.  Mol.  Biol.  8:  297. 

38.  Muench,  K.H.,  and  Berg,  P.  Purification  of  transfer  RNA.  Fed.  Proc.  23,  No.  2. 


195 

39.  Norris,  AT.,  and  Berg,  P.  Purification  and  properties  of  isoleucyl  S-RNA  synthetase 

from  Escherichia  coli..  Fed.  Proc.  23,  No.  2. 

40.  Chamberlin,  M,  and  Berg,  P.  Mechanism  of  RNA  polymerase  action: 

characterization  of  the  DNA-dependent  synthesis  of  polyadenylic  acid.  J.  Mol. 
Biol.  8:  708. 

41.  Wood,  W.B.,  and  Berg,  P.  Influence  of  DNA  secondary  structure  on  DNA- 

dependent  polypeptide  synthesis.  J.  Mol.  Biol.  9:  452. 

42.  Norris,  A.T.  Mechanism  of  aminoacytl  RNA  synthesis:  Studies  with  isolated 

aminoacyl  adenylate  complexes  of  isoleucyl  RNA  synthetase.  PNAS  52:  330. 

43.  Berg,  P.,  and  Chamberlin,  M.  On  the  transcription  of  DNA  sequences  by  RNA 

polymerase.  Bull.  Soc.  Chim.  Biol.  46:  1427. 

1965 

44.  Berg,  P.,  Kornberg,  R.D.,  Fancher,  H.,  and  Dieckmann,  M.  Competition  between 

RNA  polymerase  and  DNA  polymerase  for  the  DNA  template.  Biochem.  Biophys. 
Res.  Comm.  18:  932. 

45.  Calendar,  R.,  and  Berg,  P.  Tryosyl  RNA  synthetases  from  E.  coli  and  B.  subrilis. 

Fed.  Proc.  24:  490. 

46.  Muench,  K.H.  Resolutions  of  tRNA  on  hydroxylapatite  columns.  Fed.  Proc.  24: 

1959. 

47.  Jackson,  J.F.,  Kornberg,  R.D.,  Berg,  P.,  RajBhandary,  U.L.,  Stuart,  A.,  Khorana, 

H.G.  and  Kornberg,  A.  On  the  heterogeneity  of  the  deoxyribonucleic  acid 
associated  with  crystalline  yeast  cytochrome  b2-  Biochim.  Biophys.  Acta  108:  243. 

1966 


48.  Calendar,  R.  and  Berg,  P.  Tyrosyl  tRNA  synthetase  from  E.  coli,.  In  Procedures  in 

Nucleic  Acid  Research  (Cantoni  and  Davies,  eds.).  Harper  and  Row,  Inc.,  New 
York,  p.  384. 

49.  Baldwin,  A.N.  and  Berg,  P.  Purification  and  properties  of  isoleucyl  RNA  synthetase 

from  E.  coli .  In  Procedures  in  Nucleic  Acid  Research  (Cantoni  and  Davies,  eds.). 
Harper  and  Row,  Inc.,  New  York,  p.  400. 

50.  Baldwin,  A.N.,  and  Berg,  P.  Purification  and  properties  of  isoleucyl  ribonucleic  acid 

synthetase  from  E.  coli.  J.  Biol.  Chem.  241:  831. 

51.  Baldwin,  A.N.,  and  Berg,  P.  Transfer  ribonucleic  acid-induced  hydrolysis  of 

valyladenylate  bound  to  isoleucyl  ribonucleic  acid  synthetase.  J.  Biol.  Chem.  241: 
839. 

52.  Muench,  K.H.,  Berg,  P.  Fractionation  of  transfer  ribonucleic  acid  by  gradient 

partition  chromatography  on  sephadex  columns.  Biochem.  5:  970. 


196 

53.  Muench,  K.  H.  and  Berg,  P.  Resolution  of  aminoacyl  transfer  RNA  by 
hydroxylapatite  chromatography.  Biochem.  5:  982. 

54.  Calendar,  R.  and  Berg,  P.  Purification  and  physical  characterization  of  tyrosyl 

ribonucleic  acid  synthetases  from  E.  coli  and  Bacillus  sublilis.  Biochem.  5:  1681 . 

55.  Calendar,  R.  and  Berg,  P.  The  catalytic  properties  of  tyrosyl  ribonucleic  acid 

synthetases  from  E.  coli  and  Bacillus  subrilis.  Biochem.  5:  1690. 

56.  Carbon,  J..,  Berg,  P.  and  Yanofsky,  C.  Studies  of  missense  suppression  of  the 

tryptophan  synthetase  A-protem  mutant  A36.  PNAS  56:  764. 

57.  Carbon,  J.,  Berg,  P.  and  Yanofsky,  C.  Missense  suppression  due  to  a  genetically 

altered  tRNA.  Cold  Spring  Harbor  Symp.  31:  487. 

58.  Jones.  O.W.  and  Berg,  P.  Studies  on  the  binding  of  RNA  polymerase  to 

polynucleotides.  J.  Mol.  Biol.  22:  199. 

1967 

59.  Calendar,  R.  and  Berg,  P.  D-tyrosyl  RNA:  Formation,  hydrolysis  and  utilization  for 

protein  synthesis.  J.  Mol.  Biol.  26:  39. 

60.  Yarus,  M.  and  Bers.  P.  Recognition  of  tRNA  by  aminoacyl  tRNA  synthetases.  J. 

Mol.  Biol.  28:  479. 

61.  Slapikoff,  S.  and  Berg,  P.  Mechanism  of  ribonucleic  acid  polymerase  action.  Effect 

of  nearest  neighbors  on  competition  between  uridine  triphosphate  and  uridine 
tnphosphate  analogs  for  incorporation  into  ribonucleic  acid.  Biochem.  6:  3654. 


62.  Jones,  O.W.,  Dieckmann,  M.,  and  Berg,  P.  Ribosome-induced  dissociation  of  RNA 

from  an  RNA  polymerase  DNA-RNA  complex.  J.  Mol.  Biol.  31:  177. 

63.  Reid.  P.,  and  Berg,  P.  T4  bacteriophage  mutants  suppressible  by  a  missense 

suppressor  which  inserts  glycine  in  place  of  arginine  for  the  codon  AGA.  J.  Virol. 
2:  905. 

1969 

64.  Hill,  C.W.,  Foulds,  J.,  Soil,  L.,  and  Berg,  P.  Instability  of  a  missense  suppressor 

resulting  from  a  duplication  of  genetic  material.  J.  Mol.  Biol.  39:  563. 

65.  laccarino,  M.,  and  Berg,  P.  The  requirement  of  sulfhydryl  groups  for  the  catalytic 

and  tRNA  recognition  functions  of  isoleucyl-tRNA-synthetase.  J.  Mol.  Biol.  42: 
151. 

66.  Yarus,  M.  and  Berg,  P.  Recognition  of  tRNA  by  isoleucyl-tRNA  synthetase.  II. 

Effect  of  substrates  on  the  dynamics  of  tRNA-enzyme  interaction.  J.  Mol.  Biol. 
42:  171. 

67.  Hill,  C.W.,  Schiffer,  D.,  and  Berg,  P.  Transduction  of  merodiploidy:  Induced 

duplication  of  recipient  genes.  J.  Bact.  99:  274. 


197 


68.  Soil,  L.,  and  Berg,  P.  Recessive  lethais:  A  new  class  of  nonsense  suppressors  in 

Escherichia  coli .  PNAS  63:  392. 

69.  Soil,  L.,  and  Berg,  P.  Recessive  lethal  nonsense  suppressor  in  Escherichia  coli 

which  inserts  glutamine.  Nature  233:  1340. 


70.  Yarus,  M,  and  Berg,  P.  On  the  properties  and  utility  of  a  membrane  filter  assay  in 

the  study  of  isoleucyl-tRNA  synthetase.  Analytical  Biochem  35:  450. 

71.  Primakoff,  P.,  and  Berg,  P.  Stringent  control  of  transcription  of  phage080psu3.  Cold 

Spring  Harbor  Symposia  on  Quantitative  Biology  35:  391. 

72.  Folk,  W.R.,  and  Berg,  P.  Isolation  and  partial  characterization  of  Escherichia  coli 

mutants  with  altered  glycyl  transfer  ribonucleic  acid  synthetases.  J.  Bact.  102:  193. 

73.  Folk,  W.R.,  and  Berg,  P.  Characterization  of  altered  forms  of  glycyl  transfer 

ribonucleic  acid  synthetase  and  the  effects  of  such  alterations  on  aminoacyl  transfer 
ribonucleic  acid  synthesis  in  vivo.  J.  Bact.  102:  204. 

74.  Arndt,  D.,  and  Berg,  P.  Isoleucyl  transfer  ribonucleic  acid  synthetase  is  a  single 

polypeptide  chain.  J.  Biol.  Chem.  245:  665. 

75.  Cuzin,  F.,  Vogt,  M.,  Dieckmann,  M.,  and  Berg,  P.  Induction  of  virus  multiplication 

in  3T3  cells  transformed  by  a  thermosensitive  mutant  of  polyoma  virus.  II. 
Formation  of  oligomeric  polyoma  DNA  molecules.  J.  Mol.  Biol.  47:  317. 

76.  Ostrem,  D.L..  and  Berg,  P.  Glycyl-tRNA  synthetase:  An  oligomeric  protein 

containing  dissimilar  subunits.  PNAS  67:  1967. 

1971 

77.  Berg,  P.  The  viral  genome  in  transformed  cells.  Proc.  Roy.  Soc.  Lond.  B.  177:  65. 

78.  Arndt-Jovin,  D.J.,  and  Berg,  P.  Quantitative  binding  of  125]-concanavaJin  A  to 

normal  and  transformed  cells.  J.  Virol.  8:  716. 

79.  laccanno,  M.,  and  Berg,  P.  Isoleucine  auxotrophy  as  a  consequence  of  a 

mutationally  altered  isoleucyl-transfer  ribonucleic  acid  synthetase.  J.  Bact.  105: 

527. 

80.  Folk,  W.  Molecular  Weight  of  Escherichia  coli  glutaminyl  transfer  ribonucleic  acid 

synthetase,  and  isolation  of  its  complex  with  glutamine  transfer  ribonucleic  acid. 
Biochem  10:  1728. 

8 1 .  Folk,  W.R.,  and  Berg,  P.  Duplication  of  the  structural  gene  for  glycyl-transfer  RN  A 

synthetase  in  Escherichia  coli.  J.  Mol.  Biol.  58:  595. 

72 


82.  Folk,  W.R.,  and  Yaniv,  M.  Coding  properties  and  nucleotide  sequence.  Sequences 
of  E.  coli  glutamine  tRNAs.  Nature  New  Biology  237:  165. 


198 


83.  Jackson,  D.A.,  Symons,  R.H.,  and  Berg,  P.  Biochemical  method  for  inserting  new 

genetic  information  into  DNA  of  Simian  Virus  40:  Circular  SV40  DNA  molecules 
containing  lambda  phage  genes  and  the  galactose  operon  of  Escherichia  coli. 
PNAS  69:  2904. 

84.  Morrow,  J.,  and  Berg,  P.  Cleavage  of  Simian  Virus  40  DNA  at  a  unique  site  by  a 

bacterial  restriction  enzyme.  PNAS  69:  3365. 

1973 

85.  Beard,  P.,  Morrow,  J.F.,  and  Berg,  P.  Cleavage  of  circular  superhelical  Simian  Virus 

40  DNA  to  a  linear  duplex  by  S\  nuclease.  J.  Virol.  12:  1303. 

86.  Berg,  P.  Suppression:  A  subversion  of  genetic  decoding.  Harvey  Lectures  67:  247. 

87.  Morrow,  J.F.,  and  Berg,  P.  Location  of  the  T4  gene  32  protein  binding  site  on 

Simian  Virus  40  DNA.  J.  Virol.  12:  1631. 

88.  Morrow,  J.F.,  Berg,  P.,  Kelly,  J.K.,  and  Lewis,  A.M.  Mapping  of  Simian  Virus  40 

early  functions  on  the  viral  chromosome.  J.  Virol.  12:  653. 

1974 

89.  Beard,  P.,  and  Berg,  P.  A  convenient  micro  method  for  the  quantitation  of  closed 

circular  deoxyribonucleic  acid.  Biochem.  13:  2410. 

90.  Berg,  P.,  Baltimore,  D.,  Boyer,  H.,  Cohen,  S.,  Davis,  R.,  Hogness,  D.,  Nathans,  D., 

Roblm,  R.,  Watson,  J.,  Weissman,  S.,  and  Zinder,  N.  Potential  biohazards  of 
recombinant  DNA  molecules.  Science  185:  303. 

91.  Mertz,  J.,  Carbon,  J.,  Herzberg,  M.,  Davis,  R.,  and  Berg,  P.  Isolation  and 

characterization  of  individual  clones  of  Simian  Virus  40  mutants  containing 
deletions,  duplications  and  insertions  in  their  DNA.  Cold  Spring  Harbor  Symp. 
Quant.  Biol.  39:  69  and  in  Viral  Transformation  and  Endogenous  Viruses  (Kaplan, 
A.S.,  Ed.).  Academic  Press,  New  York,  1974. 

92.  Mertz,  J.,  a~nd  Berg,  P.  Defective  Simian  Virus  40  genomes:  Isolation  and  growth  of 

individual  clones.  Virology  62:  1 12. 

93.  Mertz,  J.,  and  Berg,  P.  Viable  deletion  of  Simian  Virus  40:  Selective  isolation  by 

means  of  a  restriction  endonuclease  from  Hemophilus  parainfluenzae.  PNAS  71: 
4879. 

94.  Ostrem,  D.,  and  Berg,  P.  Glycyl  transfer  ribonucleic  acid  synthetase  from  E.  coli: 

Purification,  properties,  and  substrate  binding.  Biochemistry  13:  1338. 

95.  Shenk,  T.,  Rhodes,  C,  Rigby,  P.,  and  Berg,  P.  Mapping  of  mutational  alterations  in 

DNA  in  S]  nuclease:  The  location  of  deletions,  insertions  and  temperature- 
sensitive  mutations  in  SV40.  Cold  Spring  Harbor  Symp.  Quant.  Biol.  39:  61. 

96.  Yaniv,  M.,  Folk,  W.,  Berg,  P.,  and  Soil,  L.  A  single  mutational  modification  of  a 

tryptophan-specific  transfer  RNA  permits  aminoacylation  by  glutamine  and 
translation  of  the  codon  UAG.  J.  Mol.  Biol.  86:  245. 


199 


1975 

97.  Berg,  P.,  Baltimore,  D.,  Brenner,  S.,  Roblin,  R.O.  and  Singer,  M.  Asilomar 

conference  on  recombinant  DNA  molecules.  Science  188:  991. 

98.  Berg,  P.,  Baltimore,  D.,  Brenner,  S.,  Roblin,  R.O.  and  Singer,  M.  Summary 

statement  of  the  Asilomar  Conference  on  Recombinant  DNA  Molecules.  PNAS 
72:  1981-1984. 

99.  Carbon,  J.,  Shenk,  T.E.,  and  Berg,  P.  Construction  in  vitro  of  mutants  of  simian 

virus  40:  Insertion  of  a  poly(dA.dT)  segment  at  the  Hemophilus  parainfluenza  II 
restriction  endonuclease  cleavage  sites.  J.  Mol.  Biol.98:  1. 

100.  Griffith,  J.,  Dieckmann,  M.,  and  Berg,  P.  Electron  microscop;e  localization  of  a 

protein  bound  near  the  origin  of  Simian  Virus  40  DNA  replication.  J.  Virol.  15: 
167-172. 

101 .  Carbon,  J.,  Shenk,  T.E.,  and  Berg,  P.  Biochemical  prodcedure  for  production  of 

small  deletions  in  simian  virus  40  DNA.  PNAS  72:  1392. 

102.  DePamphilis,  M.L.,  Beard,  P.,  and  Berg,  P.  Synthesis  of  superhelical  simian  virus 

40  deoxyribonucleic  acid  in  cell  lysates.  J.  Biol.  Chem.  250:  4340. 

103.  DePamphilis,  M.L.,  and  Berg,  P.  Requirement  of  a  cytoplasmic  fraction  for 

synthesis  of  SV40  deoxyribonucleic  acid  in  isolation  nuclei.  J.  Biol.  Chem.  250: 

4348. 

104.  Shenk,  T.E.,  Rhodes,  C,  Rigby,  P.W.J.,  and  Berg,  P.  Biochemical  method  for 

mapping  mutational  alterations  in  DNA  with  SI  nuclease:  The  location  of 
deletions  and  temperature-sensitive  mutations  in  simian  virus  40.  PNAS  72:  989. 


1976 

105.  Berg.  P.  From  enzyme  chemistry  to  genetic  manipulation.  In  Reflections  on 

Biochemistry.  Pergamon  Press,  New  York,  p.  253. 

106.  Berc.  P.,  Singer,  M.  Seeking  wisdom  in  recombinant  DNA  research.  Fed.  Proc.  35: 

2542. 

107.  Cole,  C.,  Landers,  T.,  Goff,  S.,  Manteuil-Brutlag,  S.,  Berg,  P.  Deletion  mutants  of 

SV40.  Abstract  from  SV40,  Polyoma  and  Adenoviruses,  proceedings  of  the 
Tumor  Virus  Meeting  August  18-20,  Cold  Spring  Harbor,  New  York,  p.  1. 

108.  Goff,  S.,  Berg,  P.  Construction,  propagation  and  expression  of  a  hybrid  virus 

containing  SV40  and  lambda  DNA  segments.  Abstract  from  SV40,  Polyoma  and 
Adenoviruses,  proceedings  of  the  Tumor  Virus  Meeting  August  18-20,  Cold 
Spring  Harbor,  New  York,  p.  9. 

109.  Goff,  S.,  Berg,  P.  Construction  of  hybrid  viruses  containing  SV40  and  X  phage 

DNA  segments  and  their  propagation  in  cultured  monkey  cells.  Cell  9:  695. 


200 

]  10.  Shenk,  T.E.,  Berg,  P.  Isaolation  and  propagation  of  a  segment  of  the  simian  virus  40 
genome  containing  the  origin  of  DNA  replication.  PNAS  73:  1513. 

111.  Shenk,  T.E.,  Carbon,  J.,  Berg,  P.  Construction  and  analysis  of  viable  deletion 

mutants  of  simian  virus  40.  J.  Virol.  18:  664. 

1 12.  Shishido,  K.,  Berg,  P.  Restriction  endonuclease  from  Haemophilus  gallinarum 

(Hgal)  cleaves  polyoma  DNA  at  four  locations.  J.  Virol.  18:  793. 

1 13.  Wilson,  J.H.,  DePamphilis,  M.,  Berg,  P.  Simian  Virus  40-permissive  cell 

interactions:  Selection  and  characterization  of  spontaneously  arising  monkey  cells 
that  are  resistant  to  Simian  Virus  40  infection.  J.  Virol.  20:  391. 

1977 

1 14.  Berg,  P.  Biochemical  pastimes  .  .  .  and  future  times.  The  Eighth  Feodor  Lynen 

Lecture.  In  Molecular  Cloning  of  Recombinant  DNA.  Academic  Press,  p.  1. 

115.  Berg,  P.  Mapping  the  mammalian  genome.  Forum  on  Recombinant  DNA 

Research,  National  Academy  of  Sciences,  p.  62. 

1 16.  Berg,  P.  Recombinant  DNA  research  can  be  safe.  TIBS  2:  1. 

1 17.  Berg,  P.  Genetic  engineering:  Challenge  and  responsibility.  AMBIO  6:  253. 

118.  Christiansen,  G.,  Landers,  T.,  Griffith,  J.,  Berg,  P.  Characterization  of  components 

released  by  alkali  disruption  of  Simian  Virus  40.  J.  Virol.  21:  1079. 

1 19.  Cole,  C,  Landers,  T.,  Goff,  S.  P.,  Brutlag,  S.M.,  Berg,  P.  Physical  and  genetic 

characterization  of  deletion  mutants  of  Simian  Virus  40  constructed  in  vitro.  J. 
Virol.  24:  277. 

120.  DNA  segments  and  their  propagation  in  cultured  monkey  cells.  In  Recombinant 

Molecules:  Impact  on  Science  and  Society  (Beers,  R.F.  and  Bassett,  E.G.,  eds.). 
Raven  Press,  p.  285. 

121.  Goff,  S.P.,  Berg,  P.  Structure  and  formation  of  circular  dimers  of  Simian  Virus  40 

DNA.  J:  Virol.  24:  295. 

122.  Rigby,  P.W.J.,  Dieckmann,  M.,  Rhodes,  C.,  Berg,  P.  Labeling  deoxyribonucleic 

acid  to  high  specific  activity  in  vitro  by  nick  translation  with  DNA  polymerase  I. 
J.Mol.Biol.  113:237. 

123.  Villarreal,  L.,  Berg,  P.  Hybridization  in  situ  of  SV40  plaques:  Detection  of 

recombinant  SV40  virus  carrying  specific  sequences  of  nonviral  DNA.  Science 
196:  183. 

1978 

124.  Bouck,  N.,  Beales,  N.,  Shenk,  T.,  Berg,  P.,  Di  Mayorca,  G.  New  region  of  the 

Simian  Virus  40  genome  required  for  efficient  viral  transformation.  PNAS  75: 
2473. 


201 

125.  Crawford,  L.V.,  Cole,  C.N.,  Smith,  A.E.,  Paucha,  E.,  Tegtmeyer,  P.,  Rundell,  K., 

Berg,  P.  Organization  and  expression  of  early  genes  of  Simian  Virus  40.  PNAS 
75:  117. 

126.  Goff,  S.P.,  Berg,  P.  Excision  of  DNA  segments  introduced  into  cloning  vectors  by 

the  poly(dA  dT)  joining  method.  PNAS  75:  1763. 

127.  Goff,  S.P.,  Rambach,  A.  Sstl:  A  restriction  endonuclease  from  Streptomyces  sp. 

Stanford.  Gene  13:  347. 

128.  Hsu,  M.-T.,  Berg,  P.  Altering  the  specificity  of  restriction  endonuclease:  Effect  of 

replacing  Mg2+  with  Mn2+.  Biochemistry  17:  131. 

129.  Rigby,  P.W.J.,  Berg,  P.  Does  Simian  Virus  40  DNA  integrate  into  cellular  DNA 

during  productive  infection?  J.  Virol.  28:  475. 


130.  Villarreal,  L.,  White,  R.  and  Berg,  Paul.  Mutational  alterations  within  the  SV40 

leader  segment  generate  altered  16S  and  19S.  J.  Virol.  29,  209-219. 

131.  Mulligan,  R.C.,  Howard,  B.H.  and  Berg,  Paul.  Synthesis  of  rabbit  (3-globin 

recombinant  genome.  Nature  277,  108-1 14. 

132.  Contreras,  R.,  Cole,  C,  Berg,  Paul  and  Fiers,  W.  Nucleotide  sequence  analysis  of 

two  simian  virus  40  mutants  with  deletions  in  the  late  region  of  the  genome.  J. 
Virol.  29,  789-793. 

133.  Volckaert,  G.,  Feunteun,  J.,  Crawford,  L.V.,  Berg,  Paul  and  Fiers,  W.  Nucleotide 

sequence  deletions  within  the  coding  region  for  small-t  antigen  of  simian  virus  40. 
J.  Virol.  30,  674-682. 

134.  Cole,  C.,  Crawford,  L.V.  and  Berg,  Paul.  Simian  virus  40  mutants  with  deletions  at 

the  3'  end  of  the  early  region  are  defective  in  adenovirus  helper  function.  J.  Virol. 
30,683-691. 

135.  Van  Heuverswyn,  H.,  Cole,  C.,  Berg,  Paul  and  Fiers,  W.  Nucleotide  sequence 

analysis  of  two  simian  virus  40  mutants  with  deletions-in  the  region  coding  for  the 
carboxyl  terminus  of  the  T  antigen.  J.  Virol.  30,  936-941. 

136.  Goff,  S.P  and  Berg,  Paul.  Construction,  propagation  and  expression  of  simian  virus 

40  recombinant  genomes  containing  the  Escherichia  coli  gene  for  thymidine  kinase 
and  a  Saccharomyces  cerevisae  gene  for  tyrosine  transfer  RNA.  J.  Mol.  Biol.  133, 
359. 

137.  Magnusson,  G.  and  Berg,  Paul.  Construction  and  analysis  of  viable  deletion  mutants 

of  polyoma  virus.  J.  Virol.  32,  523-529. 

138.  Mulligan,  R.C.,  Howard,  B.H.  and  Berg,  Paul.  Synthesis  of  rabbit  P  globin  in 

cultured  monkey  kidney  cells  infected  with  SV40  P  globin  recombinant  genomes. 
ICN  UCLA  Symposia  on  Eucaryotic  Gene  Regulation,  Academic  Press  Inc. 


202 

139.  Berg,  Paul.  The  physical  and  genetic  organization  of  a  viral  genome.  Crit.  Rev. 

Biochem.  7,  75-82. 

1980 

140.  Mark,  D.F.  and  Berg,  Paul.  A  third  splice  site  in  SV40  early  mRNA.  Cold  Spring 

Harbor  Symp.  Quant.  Biol.  Vol.  XLIV,  55-62. 

141.  Mulligan,  R.C.,  White  R.T.  and  Berg,  Paul.  Formation  of  p-globin  following 

infection  with  recombinants  containing  rabbit  (3-globin  cDNA  at  different  locations 
of  SV40s  late  region.  Miami  Winter  Symposium,  201-216. 

142.  Buchmann,  A.R.,  Burnett,  L.  and  Berg,  Paul.  The  nucleotide  sequence.  In  Tumor 

Viruses,  2nd  Ed.  (J.  Tooze,  Ed.)  Cold  Spring  Harbor  Monogr.  Ser.  Wb,  799-829. 

143.  Mulligan,  R.C.  and  Berg,  Paul.  Expression  of  a  bacterial  gene  in  mammalian  cells. 

Science  209,  1422-1427. 

144.  Fraley,  R.,  Subramani,  S.,  Berg,  Paul  and  Papahadjopoulos,  D.  Introduction  of 

liposome-encapsuled  SV40  DNA  into  cells.  J.  Biol.  Chem.  255,  10431-10435. 

s: 


145.  Mulligan,  R.C.  and  Berg,  Paul.  Selection  for  animal  cells  that  express  the 

Escherichia  coli  gene  coding  for  xanthine-guanine  phosphoribosyl  transferase. 
Proc.  Natl.  Acad.  Sci.  USA  78,  2072-2076. 

146.  Mulligan.  R.C.  and  Berg,  Paul.  Factors  governing  the  expression  of  a  bacterial  gene 

in  mammalian  cells.  Mol.  Cell.  Biol.  1,  449-459. 

147.  Berg,  Paul.  Dissections  and  reconstructions  of  genes  and  chromosomes.  Bioscience 

Reports  1,  269-287. 

148.  Berg.  Paul.  Dissections  and  reconstructions  of  genes  and  chromosomes.  Science 

213,  246. 

149.  Berg,  Paul.  Zerlegung  und  Rekonstruktion  von  Genen  und  Chromosomen. 

Angewandte  Chemie  93,  10,  885-893. 

150.  Subrarruni,  S.,  Mulligan,  R.C.  and  Berg,  Paul.  Expression  of  the  mouse 

dihydrofolate  reductase  cDNA  in  simian  virus  40  vectors.  Mol.  Cell.  Biol.  9,  854- 
864. 

151.  Lee,  F.,  Mulligan,  R.C.,  Berg,  Paul  and  Ringold,  G.  Glucorticoids  regulate 

expression  of  dihydrofolate  reductase  cDNA  in  mouse  mammary  tumor  virus 
chimeric  plasmids.  Nature  294,  228-232. 

152.  Southern,  P.J.,  Howard,  B.H.  and  Berg,  Paul.  Construction  and  characterization  of 

SV40  recombinants  with  (3-globin  cDNA  substitutions  in  their  early  regions.  J. 
Mol.  Appl.  Gen.  1,  177-190. 

1982 


203 

153.  Okayama,  Hiroto  and  Berg,  Paul.  High  efficiency  cloning  of  full  length  cDNA. 

Molec.  and  Cell.  Biol.  2,  161-170. 

154.  White,  R.T.,  Villarreal,  L.P.  and  Berg,  Paul.  Simian  virus  40  rabbit  (3-globin 

recombinants  lacking  late  mRNA  splice  sites  express  cytoplasmic  RNAs  with 
altered  structures.  J.  Virol.  42,  262-274. 

155.  Southern,  P.J.  and  Berg,  Paul.  Transformation  of  mammalian  cells  to  antibiotic 

resistance  with  a  bacterial  gene  under  control  of  the  SV40  early  region  promoter. 
J.  Mol.  &  Applied  Gen.  1,  327-341. 

156.  Fromm,  M.  and  Berg,  Paul.  Deletion  mapping  of  DNA  regions  required  for  SV40 

early  region  promoter  function  in  vivo.  J.  Mol.  &  Appl.  Gen.,  1,  457-48 1 . 

157.  Canaani,  D.  and  Berg,  Paul.  Regulated  expression  of  human  interferon  b]  gene  after 

transduction  into  cultured  mouse  and  rabbit  cells.  Proc.  Natl.  Acad.  Sci.  USA  79, 
5166-5170. 

158.  Goodman,  H.M.,  Berg,  P.,  Clark,  S.,  Cordell,  B.,  Diamond,  D.,  Nyugen-Huu,  C, 

Kan,  Y.W.,  Lebo,  R.V.  Structure,  evolution  and  expression  of  mammalian  insulin 
genes.  Mol.  Gen.  Neuroscience. 

1983 

159.  Okayama,  Hiroto  and  Berg,  Paul.  A  cDNA  cloning  vector  that  permits  expression 

of  cDNA  inserts  in  mammalian  cells.  Mol.  &  Cell.  Biol.  3,  2,  280-289. 

160.  Nicolas,  Jean-Francois  and  Berg,  Paul.  Regulation  of  expression  of  genes 

transduced  into  embryonal  carcinoma  cells.  Teratocarcinoma  Stem  Cells,  Cold 
Spring  Harbor. 

161 .  Oi,  V.T.,  Morrison,  S.L.  ,  Herzenberg,  L.A.  and  Berg,  P.  Immunoglobulin  gene 

expression  in  transformed  lymphoid  cells.  PNAS,  USA,  80,  825-829. 

162.  Fromm,  M.  and  Berg,  P.  Transcription  in  vivo  from  SV40  early  promoter  deletion 

mutants  without  repression  by  large  T  antigen.  J.  Mol.  Appl.  Gen.,  2,  127-135. 

163.  Fromm,  M.  and  Berg,  Paul.  SV40  early  and  late  region  promoter  function  are 

enhanced  by  the  72  base  pair  repeat  inserted  at  distant  locations  and  inverted 
orientations.  Mol.  and  Cell.  Biol.  3,  991-999. 

164.  Jolly,  D.J.,  Okayama,  H.,  Berg,  P.,  Esty,  A.C.,  Filpula,  D.,  Bohlen,  P.,  Johnson, 

G.G.,  Shively,  J.E.,  Hunkapillar,  T.,  and  Friedmann,  T.  Isolation  and 
characterization  of  a  full-length  expressible  cDNA  for  human  hypoxanthine 
phosphoribosyl  transferase.  PNAS,  USA,  80,  477-481. 

165.  Subramani,  S.  and  Berg,  P.  Homologous  and  non  homologous  recombination  in 

monkey  cells.  Mol.  &  Cell.  Biol.  3,  1040-1052. 

1984 

166.  Buchman,  A.R.,  Fromm,  M.  and  Berg,  P.  Complex  regulation  of  Simian  Virus  40 

early-region  transcription  from  different  overlapping  promoters.  Molec.  and  Cell 
Biol.  4,  1900-1914. 


204 

167.  Buchman,  A.R.  and  Berg,  P.  Unusual  regulation  of  Simian  Virus  40  early-region 

transcription  in  genomes  containing  two  origins  of  DNA  replication.  Molec.  and 
Cell.  Biology  4,  1915-1928. 

168.  Smith,  A.J.H.  and  Berg,  Paul.  Homologous  recombination  between  defective  neo 

genes  in  mouse  3T6  cells.  Cold  Spring  Harbor  Laboratory,  Vol.  XLIX. 

169.  Chin,  D.J.,  Gil,  G.,  Russell,  D.W.,  Liscum,,  L.,  Luskey,  K.L.,  Basu,  S.K.,  Okayama, 

H.,  Berg,  P.,  Goldstein,  J.L.  and  Brown,  M.S.  Nucleotide  sequence  of  3-hydroxy- 
3-methyl-glutaryl  coenzyme  A  reductase,  a  glycoprotein  of  endoplasmic  reticulum. 
Nature  308,  No.  5960,  613-617. 

1985 

170.  Naumovski,  L.,  Chu,  Gilbert,  Berg,  Paul  and  Friedberg,  E.G.  RAD3  gene  of 

Saccharomyces  cerevisiae:  nucleotide  sequence  of  wild-type  mutant  alleles, 
transcript  mapping,  and  aspects  of  gene  regulation.  Molec.  and  Cell.  Biol.  5:  17- 
26. 

171.  Okayama,  H.  and  Berg,  P.  Bacteriophage  lambda  vector  for  transducing  a  cDNA 

clone  library  into  mammalian  cells.  Molec.  &  Cell.  Biol.  5,  1 136-1 142. 

172.  Chu,  G.  and  Berg,  P.  Rapid  assay  for  detection  of  Escherichia  coli  xanthine-guanine 

phosphoribosyltransferase  activity  in  transduced  cells.  Nucleic  Acids  Research  13A 
2921-2930. 

173  Barsoum,  J.  and  Berg,  P.  SV40  minichromosomes  contain  torsionally  strained  DNA 
molecules.  Molec.  and  Cell.  Biol.  5,  3048-3057. 

174.  Dennis,  E.  and  Berg,  P.  Transcription  from  a  plant  gene  promoter  animal  cells. 
Nucleic  Acids  Research  13,  No.  22,  7945-  7957. 


175.  Canaani,  D.,  Naiman,  T.,  Teitz,  T.  and  Berg,  P.    Immortalization  of  xeroderma 

pigmentosum  cells  by  simian  virus  40  DNA  having  a  defective  origin  of  DNA 
replication.  Somatic  Cell  and  Molec.  Genetics  12,  No..  1,  13-20. 

176.  Kadesch,  T.  and  Berg,  P.    Effects  of  the  position  of  the  simian  virus  40  enhancer  on 

the  expression  of  multiple  transcription  units  in  a  single  plasmid.  Molec.  and  Cell. 
Biol.  6,  2593-2601. 

177.  Thelander,  L.  and  Berg,  P.  Isolation  and  characterization  of  full-length  cDNA 

clones  encoding  the  Ml  and  M2  subunits  of  mouse  ribonucleotide  reductase. 
Molec.  and  Cell.  Biol.  6,  3433-3442. 

178.  Peabody,  D.  and  Berg,  P.  Termination-reinitiation  occurs  in  the  translation  of 

mammalian  cell  mRNAs.  Molec.  and  Cell.  Biol.  6,  2695-2703. 

179.  Peabody,  D.,  Subramani,  S.  and  Berg,  P.  The  effect  of  upstream  reading  frames  on 

translation  efficiency  in  SV40  recombinants.  Molec.  and  Cell.  Biol.  6,  2704-271 1. 


205 

Bacillus  Molecular  Genetics  and  Biotechnology  Applications.  Ed.  by  A.T. 
Ganesan  and  James  A.  Hoch,  p.  3,  Academic  Press,  Inc.,  New  York. 

180.  McPhaul,  M.  and  Berg,  P.  Formation  of  functional  asialoglycoprotein  receptor  after 

transfection  with  cDNAs  encoding  the  receptor  proteins.  Proc.  Natl.  Acad.  Sci., 
USA  83,  8863-8867. 

1987 

181.  McPhaul,  M.  and  Berg,  P.  Identification  and  characterization  of  cDNA  clones 

encoding  two  homologous  proteins  that  are  part  of  the  asialoglycoprotein  receptor. 
Molec.  and  Cell.  Biol.  7,  1841-1847. 

182.  Chu,  G.,  Hayakawa,  H.  and  Berg,  P.    Electroporation  for  the  efficient  transfection 

of  mammalian  cells  with  DNA.  Nucleic  Acids  Research,  Vol.  15,  No.  3. 

183.  Chu,  G.  and  Berg,  P.    DNA  cross-linked  by  cisplatin:  A  new  probe  for  the  DNA 

repair  defect  in  Xeroderma  Pigmentosum.  Molec.  Biol.  Med.  4,  277-290. 

1988 

184.  Reichardt,  J.  and  Berg,  P.    Cloning  and  characterization  of  a  cDNA  encoding 

human  galactose-1 -phosphate  uridyl  transferase.  Molec.  Biol.  Med.  5,  107-122. 

185.  Margolskee,  R.,  Kavathas,  P.  and  Berg,  P.    Epstein-Barr  virus  shuttle  vector  for 

stable  episomal  replication  of  cDNA  expression  libraries  in  human  cells.  Molec 
and  Cell.  Biol.  8,  2837-2847. 

186.  Buchman,  A.  and  Berg,  P.    A  comparison  of  intron-dependent  and  intron- 

independent  gene  expression.  Molec.  and  Cell.  Biol.  8,  4395-4405. 

187.  Jasin,  M.  and  Berg,  P.    Homologous  integrations  in  mammalian  cells  without  target 

selection.  Genes  &  Development  2,  1353-1363. 

1 88.  Reichardt,  J.  and  Berg,  P.    Conservation  of  short  patches  of  amino  acid  sequence 

amongst  proteins  with  a  common  function  but  evolutionarily  distinct  origins: 
implications  for  cloning  genes  and  for  structure-function  analysis.  Nucleic  Acids 
Research  16,  9017-9026. 

1990 

189.  Jasin,  M.,  Elledge,  S.J.,  Davis,  R.W.,  and  Berg,  P.  Gene  targeting  at  the  human  CD4 

locus  by  epitope  addition.  Genes  &  Development  4:   157-166. 

190.  Stuhlmann,  H.,  Dieckmann,  M.  and  Berg,  P.  Transduction  of  cellular  neo  mRNA  by 

retroviral-mediated  recombination.  J.  Virol.  64:  5783-5796. 

191.  Pontius,  B.  and  Berg,  P.  Renaturation  of  complementary  DNA  strands  mediated  by 

purified  mammalian  heterogeneous  nuclear  ribonucleoprotein  Al  protein: 
Implications  for  a  mechanism  for  rapid  molecular  assembly.  PNAS  87:  8403- 
8407. 

192.  Singer,  M.  and  Berg,  P.  Genes  and  Genomes:  A  Changing  Perspective.  University 

Science  Books,  Mill  Valley,  CA. 


206 


1991 

193.  Jessberger,  R.  and  Berg,  P.  Repair  of  deletions  and  double  strand  gaps  by 

homologous  recombination  in  a  mammalian  in  vitro  system.  Molec.  and  Cell. 
Biol.  11:  445-457. 

194.  Berg,  P.  All  our  collective  ingenuity  will  be  needed.  FASEB  J  5:  75. 

195.  Luria,  S.,  Chambers,  I.,  and  Berg,  P.  Expression  of  the  type  1  immunodeficiency 

virus  Nef  protein  in  T-cells  prevents  antigen  receptor  mediated  induction  of  TL-2 
mRNA.  PNAS  88:5326-5330. 

196.  Berg,  P.  Reverse  genetics:  its  origins  and  prospects.  Biotechnology  9:  342-344. 

197.  Pontius,  B.  and  Berg,  P.  Rapid  renaturation  of  complementary  DNA  strands 

mediated  by  cationic  detergents:  A  role  for  high-probability  binding  domains  in 
enhancing  the  kinetics  of  molecular  processes.  PNAS  88:  8237-8241. 

1992 

198  Stuhlmarm,  H.,  and  Berg,  P.  Homologous  recombination  of  copackaged  retrovirus 
RNAs  during  reverse  transcription.  J.  Virol.  April:  2378-2388. 

199.  Pontius,  B.W.  and  Berg,  P.  Rapid  assembly  and  disassembly  of  complementary 

DNA  strands  through  an  equilibrium  intermediate  state  mediated  by  Al  nhRNP 
protein.  J.  Biol.  Chem  267:  13815-13818. 

200.  Berg,  P.  and  Singer,  M.  Dealing  with  Genes:  The  Language  of  Heredity. 

University  Science  Books,  Mill  Valley,  CA.. 

1993 

201.  Jessberger,  R.,  Podust  V.,  Hiibscher,  U.  and  Berg,  P.  A  mammalian  protein  complex 

that  repairs  double-strand  breaks  and  deletions  by  recombination.  J.  Biol.  Chem. 
268:  15070-15079. 

202.  Berg,  P.  Co-chairman's  remarks:  reverse  genetics:  directed  modification  of  DNA 

for  functional  analysis.  Gene  135:  1  62-164. 


1995 

203.  Firmenich,  A.,  Elias-Arnanz,  M.,  and  Berg,  P.  A  novel  allele  of  Saccharomyces 

cerevisiae  RFA1  that  is  deficient  in  recombination  and  repair  and  suppressible  by 
RAD52.  Molec.  and  Cell.  Biol.  15(3):  1620-1631. 

204.  Zieler,  H.,  Walberg,  M.,  and  Berg,  P.  Suppression  of  mutations  in  two  5.  cerevisiae 

genes  by  the  adenovirus  El  A  protein.    Molec.  and  Cell.  Biol.  15:  3227-3237. 

205.  Hays,  S.L.,  Firmenich,  A.A.,  and  Berg,  P.  Complex  formation  in  yeast  double- 

strand  break  repair:  Participation  of  RadSl,  Rad52,  Rad55,  and  Rad57  proteins. 
PNAS  92:  6925-6929. 


209 


Paul  Berg 

Application  #10170         \_l  %»^  j 

We  are  trying  to  learn  how  viral  genes  can  alter  the  growth 
properties  of  a  cell  and  make  it  cancerous.     An  important  goal  of  our 
research  is  to  learn  how  genetic  information  carried  by  a  virus 
chromosome  can  be  integrated  into  the  genetic  machinery  of  a 
mammalian  cell  and  thereby  influence  the  properties  of  that  cell. 
To  study  this  we  are  examining  a  mutant  virus  which  is  unable  to 
transform  cells  into  tumor  cells  under  certain  conditions.     We  hope 
to  define  the  defective  function  and  to  determine  its  role  in  the  inte 
gration  process.     In  practical  terms,    if  we  could  understand  how  new 
genetic  information  can  be  inserted  and  maintained  in  a  foreign 
chromosome,   we  might  not  only  be  closer  to  the  secret  of  cancer, 
but  also  on  the  road  to  learning  how  to  modify  the  genetic  constitution 
of  cells  by  integration  of  other  types  of  DNA  molecules. 


.,  _. 


sip  uiojg  uotssnmad  saimbai  uoponpoodai  jsqimj  'Xpio  asn  aouawjai  jqj  si 


210 

' 

of  Preliminary  Planning  of  Basic  Sciences  Sub-  Committee 
of  Cancer  Center  Planning  Group 


A  strong  Basic  Sciences  Program  is  an  essential  element 
any  effective  Cancer  R.crearch  effort.     Without  the  continual  infusion 
basic  science  techniques,   knowledge, and  ideas,   the  momentum  and 
^inspiration  for  the  needed  research  developments  on  the  Cancer  Problem 
••would  soon  founder  and  dry  up.     With  this  premise,    and  indeed  strongly 
rbeld  conviction,   the  Basic  Sciences  Sub-Committee  of  the  Cancer  Center 
i. Planning  Group  has  conducted  its  preliminary  planning  by  considering 
following  three  general  questions: 


|l)  Stanford's  present  strengths  in  those  areas  of  Basic  Sciences  Research 
»*  which  are  relevant  or  bear  directly  on  the  Cancer  Problem. 

-2)  Stanford's  needs  for  expanding  ,    and  strengthening  existing  programs 

••"as  well  as  development  of  new  programs  in  various  areas    of  Cancer- 

?  related  Basic  Sciences  Research. 

i  . 

\:3)  Ways  in  which  the  Cancer  Center's  Basic  Sciences  Research  Activities 

should  be  organized  so  as  to  enhance  the  Center's  Clinical  efforts  as  well 
'  as  be  consistent   with  Medical  School's  existing   Research  and  Teaching 

Programs.  .  .. 

No  definitive  conclusions  have  been  reached  yet   on  any  of 
;  these  three  problems  but  the  following  outline  attempts  to  summarize  some  of 

our  thinking  in  these  areas.      For  clarity  and  brevity,    the  first   two 
"questions  have  been  considered  together  under  each  heading. 

1  )      Present  Strengths  and  Activities  in  Basic  Sciences  Research 
Relevant  to   the  Cancer  Problem. 


A)  Molecular  and  Regulatory  Biology  of  Animal  and  Human  Cells: 

Stanford's  emphasis  in  this  area  of  research  has  dealt     mainly 
with  bacterial  cells  and  their  viruses  but  more  recently  there  has 
been  an  expansion  of  research  with  eukaryote  systems.     In  spite 
of  Stanford's    acknowledged  and  recognized   leadership  in  this  field 
an  objective    review  of  these  activities    shows   that  it   is  the 
intensiveness  rather  than  extensiveness  that  is  our  strength. 


o 


i)  Basic  Mechanisms  and  Structures  Concerned  with  Replication, 
Transcription,    and  Translation. 

Perhaps  strongest  is  research  centered  about  the 
enzymatic  synthesis  of  nucleic  acids,   and  in  particular  in 
prokaryote  systems.     Dr.   Arthur  Kornberg  (Biochemistry) 
is  continuing  his  elegant  studies  on  enzymes  involved  in  DNA 
synthesis.     Dr.   1.  R.    Lehman  (Biochemistry)  has  been  concerned 
for  many  years  with  various  enzymes  concerned  with  DNA 


~     K> 


2. 

211 

metabolism,    and  is  currently  studying  the  enzymatic  mechanisms 
of  genetic  recombination  in  E.    coli  .     Dr.    A.  D.    Kaiser 
(Biochemistry)  has  worked  at  a  slightly  different  level  of  DNA 
synthesis  in  studying  the  processes  whereby  DNA  is  synthesized 
and  packaged  into  a  completed  virus  particle,    studies  that  have 
combined  genetic,    enzymatic,    and   electron  microscopic  approaches. 
Dr.    David  Hogness   (Biochemistry),    whose  previous  work  had  been 
on  the  genetics  of  lambda  phage,    has  focused  his  undivided  attention 
in  the  last  two  years  on  the  molecular  structure,    regulation  and 
expression  (developmental)    of  polytene     chromosomes   of 
Drosophila.     Dr.   A.    Ganasan  (Genetics)  has  been  studying   the 
process  of  DNA  replication  in  prokaryotes,    and  more  recently 
in  animal  cells,    centering  his  studies  on  the  potential  role  of 
nuclear  membrane- chromosome  complexes  in  DXA  replication. 
Dr.    David  Clayton  (Pathology)  is  concerned  with  the  synthesis 
and  function  of  complex  forms  of  DNA  associated  with 
mitochondria  of  malignant  cells.      Dr.   Paul  j  erg   (Biochemistry) 
has  made  important  discoveries  concerning  the  structure  and 
function  of  RNA  polymerase,    the  mechanism  and  regulation'  of 
ammo  acid _a.ctivation  imcf  protein  synthesis  in  prokaryote"s.     "Not 
directly  associated  witH'the "medical  school,   but'  an  active 
intellectual  input  to  the  work  going  on  .here  is  Dr.    Charles 
Yanofsky's  group  (Biological  Sciences),   whose  studies  on  the 
regulation  of  the  tryptophan  operon  in  E.    coli    has  been  pioneering 
and  one  of  the  most  germinal  in  the  field  of  regulatory   biology. 
Dr.    R.    Schimke  (Pharmacology)  has  been  concerned  for  a  number 
of  years  with  the  mechanism  and  regulation  for  continual  synthesis 
and  degradation    of  cellular  proteins  of  animal  cells.     Dr.    Oleg 
Jardetzky  (Pharmacology)  is   studying  dynamics  of  protein  folding 
and  molecular  interactions  of  DNA  and  regulatory  proteins,    spec 
ifically  the  lac  represser,    by  various  physical  techniques,    including 
NMR  spectroscopy. 

ii)  Regulatory  Processes-including  Developmental,    Environmental, 
and  Hormonal  Influences 

Dr.   Arthur  Kornberg  (Biochemistry)  has  been  studying 
the  simplified  developmental  system  of  sporulation  in  B .    subtilis  , 
a  process  that  involves  degradation  of  cytoplasmic  proteins,    and 
the  formation  of  an  essentially  "new  cell  type,    the  spore.  A  number 
of  individuals  at  Stanford  Medical  School  are  concerned  with  various 
aspects  of  regulatory  processes,    particularly  those  that  concern 
developmental  and  hormonal-controlled   processes.      Dr.    R.    Schimke 
(Pharmacology)  is  investigating      steroid  hormone    regulation  of 
cytodifferentiation  and  function  of  chick  oviduct  with  the  eventual 
goal  of  isolating  all  regulatory  components  concerned  with  specific 
protein  synthesis,    i.e.    genes,   mRNA  ,   protein  synthesis  factors, 
etc.   and  analyzing  their  response  and  behavior  with  respect  to 
developmental  and  hormonal  stimuli.     Dr.   Aronow  (Pharmacology) 
has  been  examining  the  effects  of  corticosteroids  on  animal  cells 
in  culture   with  a  view   to  defining  the  mechanism    of  that  control 
system.     Dr.   Merton  Bernfield    (Pediatrics)  is  also  interested 
in  hormonal  regulation  of  development,    and  is  specifically  studying 
the  role  of  alterations  in  isoaccepting  tRNA  species  in  the  regulation 
of  specific  protein  synthesis  in  chicken  liver.     Another  aspect  of 
his  studies  is  related  to  the  role  of  epithelial-mexenchymal 

'suop3an°D  I^P^S  3° 


212 

interactions  in  control  of  differentiation.      Dr.    Lawrence    Kedes 
(Medicine)  is   studying   the  regulation  of  histone  synthesis  in 
the  developing  sea  urchin,    and  in  particular  is  utilizing  techniques 
to  isolate  labeled  mRNA  for  hi  stone     biosynthesis,    to   map   the 
hi  stone    genes  on  the  chromosome,    and  to  study  the  activation 
of  these  genes  during  early  emryogenesis.      Dr.    Frank  Stockdale 
(Medicine)  actively  studies    the  hormonal    regulation  of  mammary 
gland  development,    and  the  molecular  events  involved  in  myogenesis 
Dr.    Stanley^jpphen  (Medicine)  has  made  important  contributions 
to  ouf~uft'de"r stan ding  of  the  mechanism  of  developme_nt_of_resistance 
to  a  number  of  antibiotics  b y ~a c qui s itfon~bf  "d ru g  resistance"  factors  . 
He,    as  well  as  Dr.    Korn's    laboratory  (Pathology),    have  made  detail 
ed    investigations  of  the  regulation  of   episomes,    replication  and 
transfer    and  its  bearing    on  drug  resistance  and  other  pharotypic 
properties.      Dr.    Eric  Shooter's  group   ha.s  had  a  strong  program 
on  analyzing   the   structure    and  function    of  the  nerve  growth  factor; 
(NGF)  ;    2\GF  is  a  protein  of  animal  origin   which  specifically 
stimulates  outgrowth  of  fibers  from  embryanic  sympathetic 
ganglia. 

An  Institute  program  in  cancer  biology  will  need 
considerable  development  in  a  number  of  areas  related  to 
macromolecular   structure  and  synthesis.      There  is    particular 
need  for  a  group  of  investigators  who  are  capable  of  studying 
the  molecular  structure  of  proteins  and  nucleic  acids,    including 
X-ray    crystallography  and  sequencing  of  both  proteins  and  nucleic 
acids.      Facilities  and  expertise  for  these  approaches,    so 
fundamental  to  an  understanding  of  regulatory  processes,    is 
totally  lacking  at  Stanford.      Other  areas  to  be  developed  include 
the  general  area  of  regulation  of  DNA  replication  in  animal 
cells.      Programs  should  also  be  developed  in  the  area  of  the 
structure  and  function  of  animal  chromosomes,    including  the 
methodology  for  isolation  of  specific  chromosomes  and  the  analysis 
of  DXA -protein  interactions  by  various  physical  and  biochemical 
techniques . 

Another  area  of  research  which  is  an  integral  part  of  a 
cancer  biology   program  and  should  be  expanded  at  Stanford 
concerns  the  structure  and  function  of  intracellular  organelles. 

CELL  ORGAXELLES 

I.    Recent  advances  in  the  biology  of  mammalian 
cells  have  shown  that  is  is  quite  feasible  to  study  the  various 
organelles  of  the  malignant  cell  as   separate  entities.     In 
particular,    cell  membranes,  microfilaments  and  mitochondria 
have  received  increasing  attention  in  the  last  few  years.     From 
these  investigations  it   is  clear  that    such  systems  are  fruitful 
areas  to  pursue  fundamental  problems  in  mammalian  cell 
Biology. 

The  specific    areas  currently  under  investigation  by  existing 
Stanford  faculty  are  as  follows: 


Xjis»Aiuft  puojirejs  'suop»n<O 
sin  man  uotssiuuad  sarmbai  uonanDoidai  jatnm j  -Xruo  asn  aauaraiai  JDI  si 


213 

genome  in  human  tumor  cell  populations  taken  at  biopsy.     This  approach  tits 
in  closely  with   the    proposed   Human  Cancer  Cell  Bank  described  in 
section  E.     In  Hayflick's  program  human  diploid  cells  will  be  used  to 
determine  whether  several  different  oncogenic  viruses  including 
feline  and  murine  sacoma  and  leukaemia  viruses  can,    under  special 
conditions,    transform  these  normal  human  cells  to  cells  having  cancer 
properties.     These  special  conditions  include: 

a)  Extremely  low  virus  multiplicities,    coupled  with  the 
addition  of  specific  immune   serum. 

b)  Transformation  of  human  diploid  cells  will  be  attempted 
utilizing  a  combination  of  very  low  level  radiation  and 
addition  of  low  multiplicities  of  several  oncogenic  RNA 
viruses  at  several  times  post-irradiation. 

c)  Transformation  of  human  diploid  cells  are  being  attempted 
using  very  low  multiplicities  of  several  different  human 
viruses  plus    reduced  temperature  of  incubation. 

d)  Attempts  to  transform  normal  human  cells  with  chemical 
carcinogens  as  a  means  of  comparing  cell  susceptibilities 
to  viral   chemical       carcinogenesis;    i.e.    the  compounds  of 
3  methylcholanthrene  and  benzo  (c  )-pyrene. 

3)  Dr.    :?aul  Berg's  group  (Biochemistry)  has  an  expanding  and  extensive 
research  program  on  the  Molecular  Biology  of  the  DNA  Tumor  Viruses  .- 
Polyoma  (PY)  SV40  and  adeno  viruses.     Their  work  is  proceeding  along 
the  following  lines: 

a)  Identification  of  viral  genetic  functions  for  both  lytic  growth 
and  cellular  transformation  leading  to  oncogenic  potential 

in  animals. 

b)  The  mechanism  and  regulation  of  expression  of  these  genes 
particularly  the  intsrplay  of  the  host  cell's  and  viral  regulatory 
systems  on  each  other;  i.e.   how  viral  genes  perturb  cell's 
growth  regulatory  machinery  and  how  cell's  regulatory  systems 

— l^m^LB__i__^_^^___im*Jt  J«'   '"l-'tl .11  mi  r  ill  irji jf ni-i.*. 

affect  readout    of  viral  gdnes. 


214 

c)  Identification  of  viral  transcripts  and  the    protein  products 
produced    in  infected  and  transferred  cells. 

d)  In  vitro    analysis  of  virus-directed  processes  in  sub-cellular 
systems;  i.e.   nuclear  and  enzyme  preparations. 

e)  Development  of  a  system  for  viral  mediated  transduction 
of  genetic  information  from  one  cell  to  another. 

4)  Recently  Dr.    George  Stark  (Biochemistry)  has  begun  a  program  to  isolate, 
characterize    and  define  the  function  of  the  T-antigen  produced  in  cells 
transformed  and  carcinogized  by  these  PY  ad'SV40  viruses. 

5)  Dr.    William  Robinson's  program   (Medicine)  is  directed  at   the  Molecular 
Biology    of  the  RXA  Tumor  Viruses  particularly  the  avian  group.      These 
are  the  best   known  and  so  far  most  intensively  studied.    Dr.    Robinson  has 
already  made  numerous  important  contributions  to  our  knowledge  of  the 
structure  of  the  viral  RNA  genome,    of  the  GSA  and  the  Type  specific 
envelope    antiger.s.     He  is  currently  studying  the  synthesis  of  these 

viral- specific  components  in  transformed  cells  as  well  as  the  nature 
and  activity  of  the  newly  discovered  RNA-dependent  DNA  polymerase. 
Quite  clearly  this   system  has  been  germinal   in  directing   attention  to 
the  possibility  that   the  RNA  tumor  viruses  may  well  be  important   for 
understanding  human  cancers.      Certainly  many  of  the  observations  with 
the  avian  system  has   had  direct  relevance  to   the  C-type  viruses  of  other 
species. 

It's  cuite  clear  that  there  is  significant   activity  now  going  on 
at  Stanford    in   this  field  of  research.     What  is  needed  is  an  expansion 
of  these  activities  particularly   for  increased  efforts    on  genetic  approaches 
to   the  RNA  Tumor  Viruses  and  for  work  with  other  mammalian  RNA  Tumor 
Viruses   (feline,    murine,    etc.  )     Even  more  gaping  in  our  inadequacy  is 
work  with  other  animal  viruses.      There  presently  is  no  substantial  work 
with  pox  viruses  of  the  reovirus  type.     All  of  these  provide  fertilization 
for  work  on  the  frank  tumor  viruses.     We  have  little    or   no   ongoing   work 
with   Herpes  virus  yet  this  class  has  recently  been   implicated  in  naturally 
occurring  human  as  well  as  animal  cancer.     As  pointed  out   in  another  section, 


215 
STANFORD  UNIVERSITY  MEDICAL  CENTER 

STANFORD.  CALIFORNIA  94305 

I  , 

\RTMENT  OF  BIOCHEMISTRY  PAUL  BERG 


Jack,  Lulu  and  Sam  Willson 
Professor  of  Biochemistry 


February  29,    1972 


Dr.    Michael  Stoker 
Imperial  Cancer  Research  Fund 
P.  O.    Box  123 
Lincoln's  Inn  Fields 
'London,    W.  C.    2,    England 

Dear  Michael: 

So  often  in  the  last  few  weeks  I've  wanted  to  write  to  you  but  an  avalanche 
of  deadlines,    from  which  I  am  only  recently  recovering,    forced  delay  after  delay. 
How  are  things  at  ICRF?     Hopefully,    science,    construction  progress  on  the  new 
labs  as  well  as  the  many  administrative  activiii.es  you  are  involved  in  are  going 
well;  Peter  Beard's   occasional  correspondence  with  people  in  the  lab  and  Bill 
Folk's  visit  brought  only  incomplete  accounts  of  the  happenings  in  London.      Right 
now  I'm  wondering  how  you're  faring  under  strictures   oi'-the  fuel-electricity 
shortage;   sounds  pretty  grirn!      Has  it  caused  hardships  in  the  labs,    particularly 
with  being  able  to  maintain  necessary  services   for  work  and  keeping  cells 
viable  and  alive0     I  suspect  that  long  ago  you  had  seen  to  ensuring  the  labora 
tories   own  power   supplies  and  therefore  immunity  from  external  exigencies. 
From  what  I  read  in  our  newspapers  I  suspect  you  probably  had   trouble 
commuting  from  Kent. 

In  the  last  few  months  things  have  gone  quite  well  here    in  the  lab.    Let 
me  fill  you  in  on  a  few  of  the  details. 

1)  We've  mastered  the  technique  of  detecting  polyoma  or  SV40  viral  DNA 
seouences  by  following  the  kinetics   of  annealing  using  S,,    an  enzyme  which 
rapidly  degrades   single- stranded  DNA  but  leaves  double- stranded  DNA  segments 
intact.      In  a  nutshell,    (I  assume  Bill  Folk  can  relay  more   specific  details  of 
the    procedure   since  he  followed  our  protocols  to  the  letter),    we   synthesize 
very  highly  p'--labeled  PY  or  SV40  DNA  using  pure  E.    coli    DNA  polymerase 
highly  purified  unlabcled  Form  I  DNA  as  template--*    four  very  hot  d-triphosphates 
(10-15  mc/i-mole)  and  traces  of  DN'ase  to  generate   random  single- strand  nicks 
in  the   supercoils.      DNA  polymerase   replicates   random  segments  of  the  DNA 
(by  "nick-translation")  thereby  generating  highly- labeled  viral  DNA  segments  . 
The  labeled  DNA  which  has  an  average  single- strand  length  of  300-500  bases 
and  a   specific  activity  of  5  -  10  x  10°     cpn-i/p-g  is  used  as  the  "probe"  for  detecting 
complementary  sequences  in  cellular  DNA's.     The  experiment  is  quite  simple 
now;  e.g.    P      -PY  DNA  is  denatured  in  the  presence  of  either  salmon-sperm 
DNA,    BHK  DNA  or  the  DNA  from  cells  abortively  or   stably   transformed  by  PY. 
Salt  is  added  to  the  appropriate  concentration  (0.  2-1.  5M  depending  on  the    rrxte 
of  annealing  wo  expect)  and  annealing  occurs  at  63°.     Samples  are  periodically 
withdrawn,    diluted  and  frozen.      When  enough  samples  have  been  taken,    they 
are  all  digested  briefly  with  S^  and  then  precipitated  with  TCA.     The  amount  of 
annealed  DNA  is  equal  to   the  amount  of  P"""- label  precipitated  by  TCA  and  re- 

t-;iinr>r!    on    thi'    filter.        At    y.r-rn   tim.=    iliics    ?_.!<'',. nT  t>ir>    irmnl    af   tl-io    «=>nd    of  tVu» 


216 

Dr.    M.    Stoker 
February  29,  1972 

Page  Two 

70-80To  of  the  DNA  is  insensitive  to  S,,    (the  remainder  is  probably  the  fraction 
of  DNA  nucleotides  which  cannot  enter  into  helical  structure  because  of  steric- 
hindrance,    or  we  believe  more  likely,    the    non- viral  DNA  sequences  carried  by 
our  polyoma  stock).     The  protocol,    computer  printouts  of  the  data  and  calculati 
as  well  as  the  photographs   show  a  typical  reconstruction  experiment  when  know 
quantities  of  cold  PY  DNA  (sheared  to  same  size  and  denatured)  is  added  to  the 
annealing  mixture.     Recently,    we've  simplified  this  procedure  so  that  samples 
are  taken  only  during  the  first  few  hours  of  annealing  and  cur  computer  prograr 
computes  the  second-order  rate  constant  (and  thereby  Cot  1/2)  from  the  initial 
points.     The  second  set  of  computer  printout  and  plots  use  only  the  data  for  the 
first  24  hours  and.  you  can  see  that   the  curves  are  linear  (2nd  order)  even  with 
the  points  for  the  first  two  hours.      The  sensitivity  seems  to  be    good  enough 
to  detect  0.  5-1  genome  equivalent  per  cell  (<  1  part  in  10°)  using  approximately 
1.  5  to  2.  5  mg  of  cell  DNA. 

Using  this  method  we:ve  begun  to  analyze  some  of  the  abortive  and  stab] 
transformants  of  EHK  we  collected  when  I  was  in  London.     Summarizing: 


c  • 


a)  p--FY  DNA  in  the  presence  of  salmon  sperm  or  3KK  DNA  anneals 
with  an  identical  Cot  1/2:  Martin  claims  that  3T3  does  contain  some  sequences 
homologous  to  SV40  but  we   shall  have  to  do  this  mar.y  time  3  before  one  can  say 
definite!-,-  that  there  is   or  is  not  a  fraction  of  a  PY  genome  equivalent  in  normal 
BHK  cells. 

b)  The  abortives  MA- S   (rnethccell)  and  SA-10   (surface-infected)  have  no 
detectable  PY   sequences.      Of  the  two  stables,    transformar.ts  we've  tested,    ST- 
and  MT-1,    only  MT-1  contains  PY-  DNA  sequences.  (MT-1  3-5  viral  equivalents 
ST-1  <    1  viral  equivalent /cell).      Consequently,    ST-1  may  be  a  spontaneous 
transfcrmant  but    it  should  be   retested.     If  you  have  another  sample  of  ST-1 
withpne    that  can  be   screen  hopefully,    one  that  could  be  tested  for  T-ag,    we'd 
like  to  retest  it. 

c)  We  have  just  finished  growing  MA-4,    MA-6  and  MA- 9  and  SA-2  as 
\VQ'.'   :-.s  one  other  stable  trarisformant  and  will  do  the  annealing  kinetics  with 
their  DNA's   shortly. 

In  view  of  Smith  _e_t_  a_L  finding  with  the  SV-3T3  abortives  it  really  is 
important  to  determine  for   sure  if  PY-BHK  abortives  really  do  not    have  PY- 
DNA  sequences  in  their  DNA.     If  there  is  a  difference  it  could  be  very  relevant 
to  the  mechanism  of  integration- excision  with  eac*.  virus.     It  seems  crucial 
therefore,    that  we  look  at  as  many  true  abortives  as  possible  to  be   sure.    I  hav> 
only  some  of  the  clones  we  picked.    Do  you  have  more?     Or  even  abortives  coll 
in  different  experiments  over  the  years?     Or  do  we  have  to  do  another  experim 
collect  a  new  set  (clones  grown  from  microcolonies  picked  from  methocell  wou 
seern  to  be  the  best  since  they  are  most  likely  abortive).    I'd  be  delighted  to 
come  over  again  to  prepare  a  fresh  lot  but  that's  not  possible  now.     Some  time 
in  September   (Renato  has  been  trying  to  talk  me  into  coming  when  he's  there  ir 
Sept.  )  would  be  possible  but  that  seems  a  long  way  off;  until  them  is  it  possible 
to  get  anv  abortively  transformed  clones,    (either  ones  we  collected  in  19"0  or 


s3Lrejqr[  XjiswAtufi  pjojirejs  'suonoanoD 


217 

Dr.M.   Stoker 
February  29,    1972 
Page  Three 

any  others  you  have)  to  look  at?     Accumulating  enough  cells  for  DNA  isolation 
is  a  limiting  factor  since  our  large-scale  growing  facilities  are  still  primitive. 
Is  it  possible  for  your  set-up  to  produce  about  5  gms  wet  weight  of  some    of  the 
clones0     If  we  could  grow  some  and  you  could  grow  others  that  might  speed 
things  up  somewhat.     Unfortunately,    although  we  are  in  good  shape  to  do  the 
hybridizations  easily  enough  we  are  in  worse  shape  to  generate  and  grow  well 
characterized  abortives. 

2)    Our  project  to  insert  new  segments  of  DNA  into  the  SV40  DNA  molecv 
has   succeeded  and  now  we  hope  to  get  to  sorrre  interesting  experiments.    Without 
'going  into  detail  here,    unique  SV40  linear  DNA  molecules   (made  by  a  single 
double- stranded  break  with  a  bacterial  restriction  enzyme  coded  for  by  the 
drug- resistance    transfer  factor  RTF-1)  have  been  derivatized  at  their  3 'OH  end 
with  either  short  runs  of  dA  or  dT  and  after  annealing  to  produce  non-covalent 
dimer  circles  they  were  covalently  joined  with  DNA  pciymerase  and  DNA  ligase 
to  produce  sealed  dimers  in  25-35fo  yield.     Using  the  same  kind  of  SV49  DNA     / 
linears  and  appropriately  derivitized  linear  DNA  from  \cygal  (mol.    wt.     6  x  10 
containing  about  4  x  1C"  daltons  of  X  DNA  and  about  2  x  10°  daltons  of  E.    coli  DN 
including  the  entire  gal  operon),    covalently  joined  hybrid  DNA  molecules  have 
been  made  in  about  20-25%  yield.     Still  to  be  completed  (this  has  run  into  more 
difficulties  than  I  anticipated)  is  the  insertion  of  a  short   piece  of  synthetic 
DNA,    dl,  G:dC     (100  base  pairs),    into  the  SV40  ring  at  the  R.  restriction  site 
and  ultimately  at  other   sites  in  the   structure.    Hopefully,    such  molecules  may 
give  us  a  way  to  map  the  genes  of  SV40. 

There  are  several  other  projects  moving  well  enough  (Peter  Beard's 
work  is  coming  along  quite  nicely)  but  more  about  these  at  some  future  occasion 
either  here,    in  London,    or  at  least  by  mail. 

I  do  lock  for'.vard  to  your  comments   on  the  question  of  the  abortives  and 
c:  course  to  hearing  something  about  your  c\vn  work  on  the  serum  factors,    as 
well  as   of  other  happenings  from  ICRF. 

Jv'y  very  best  wishes  and  regards  for  the  coming  year  to  your  family, 
and  colleagues  at  ICRF.      Please  remember  me  to  all. 

Sincerely, 


PB/1 


XiiswAiun  nionnnc  'suonosnon  mo*k  jo 


218 
STANFORD  UNIVERSITY  MEDICAL  CENTER 

STANFORD.  CALIFORNIA  94305 

IEPARTMENT  OF  BIOCHEMISTRY  PAUL  BERG 

Jack,  Lulu  and  Sam  Wilbc 
Professor  of  Biochemistry 

March  28,   1972 

Dr.   Michael  .er 

Imperial  Cancer  Research  Fund  Laboratories 

P.O.   Box  123 

Lincoln's  Inn  Fields 

London,    England 

Dear  Michael: 

Your  secretary  wrote  me  of  your  visit  to  the  States;  I  hope  that 
you  feel  welcome  enough  at  Stanford  to  let  me  know  when  you're  within 
striking  distance  of  Palo  Alto  so  that  we  can  arrange  for  a  visit.     We've 
come  a  long  way  since  your  last  visit  but  more  importantly  we'd  love 
to  see  you  again. 

It's  really  marvelous  that  you're  able  to  resurrect  more  of  the 
abortives  and  stables  and  to  send  us  cells  for  the  DNA  preps.     We  have 
trypsinized  the  cells,    quenched  with  serum  and  washed  1  time  by 
centrifugation  with  Tris- saline  and  then  frozen  the  cell  pellets.     If  we 
got  the  cells  you  indicated  that  way  it  would  be  great.     With  respect  to 
ST-1,    we  are  repeating  the  determination  with  another  batch  of  DNA  and  we'll 
do  it  with  your  cells  as  well;  that  should  give  a  pretty  definitive  answer. 
As  you  say,    if  they  remain  transformed  without  having  the  viral  DNA  that 
would  be  interesting.     Do  you  have  anti  PY-T  serum?     It  could  be  important 
to  know  if  ST-1  is  T-ag~  or  T-ag~. 

I  spoke  to  Helene  Smith  last  week  and  she  passed  on  some  additional 
information.     The  hybridizable  SV40  DNA  in  one  of  their  isolates  sediments 
with  the  cell  DNA  in  an  alkaline  sucrose  gradient  done  according  to  Sambrook 
_et  al.     She  can  detect  "no:i  infectious  SV40  DNA  in  such  cells.     Presumably 
then  it  is  "integrated".     But  in  one  of  their  clones,   the  one  which  gave  variable 
amounts  of  SV40  sequences  in  different  clonal  isolates  of  the  original  abortive, 
and  on  different  trials,    contained  no  SV40  DNA  after  growing  up  a  large  batch 
of  cells.     Very  strange  indeed.     The  abortives  that  contain  DNA  show  about 
1%  of  the  transforming  efficiency  of  normal  3T3  with. wild- type  SV40  DNA 
virus.     Didn't  you  find  that  BHK  abortives  could  be  transformed  at  normal 
efficiencies  when  challenged  with  PY? 

We  plan  to  make  highly  labeled  P      -SV40  DNA  for  use  in  detecting 
SV40  sequences.     The  .procedure  would  be  the  same  as  for  PY;  purified  SV40 
Form  I  is  used  as  template  instead  of  PY  I  in  the  DNA  polymerase  reaction 
and  that  generates  that  reagent  for  annealing.     We  could  test  Warner's  clones 
when  they're  ready. 

We've  just  had  another  set  of  interesting  results  you  might  like  to 
sauBjqn  XjMJSAiun  pwju^S  'suoposnco  FP*Js  P  waunredsQ 

,i,rttT  .tniccTiiifvf  eaTinhai  unmrmo irfoj  JOirun  J   'XlUO  3ST1  3311313131  JOJ  SI  XdOOOlOIM 


219 
Dr.   M.   Stoker 

Page  Two 
March  28,    1972 

hear.    -One  of  my  students,   John  Morrow  ,   has  been  studying  the  action  of 
the  restriction  enzyme,  Rj  (coded  for  by  the  Resistance  Transfer  Factor  I, 
RTFT,    carried  by  E.    coli)  on  SV40  DNA.     This  enzyme  has  been  purified 
by  Herb  Boyer  in  SF  who  gave  us  some  for  these  experiments.     RT  cleaves 
SV40  Form  I  DNA  (prepared  from  plaque  purified  virus)  quantitatively  to 
unit  length  linear  molecules;  the  distribution  of  lengths  (as  seen  in  EM)  is 
within  2%  that  of  the  circles.     Each  single  strand  is  unit  length  in  alkali 
so  quite  clearly  the  enzyme  makes  only  a  double- strand  cleavage  and  no 
more.     The  molecules  are  unique;  when  denatured  and  then  renatured  they 
produce  only  linear  molecules  with  the  same  length  distribution  as  the  starting 
molecules.     Had  they  been  circularly  permuted  linears  or  even  two  or 
several  types  such  denaturation- renaturation  produces  circular  molecules 
(the  result  found  with  linears  produced  by  PT  or  B- restriction  enzymes  or 
with  DNA'ase).     Further  proof  of  the  uniqueness  of  the  brea^is  the  following: 
Delius,    at  CSH,   has  found  that  T4  phage  gene  32  protein  binds  to  SV40  DNA 
Form  I  and  if  fixed  to  the  DNA  with  glutaraldehyde  and  then  spread  on  grids 
for  EM  inspection  one  obtains  molecules  with  a  single    "  bubble.  " 


If  such  molecules  are  treated  with  RT  one  obtains  linear  molecules  with 
a  bubble  at  only  one  location; 


about  40%  in  from  one  end.     Thus  we  can  differentiate  one  end  from  the  other. 
Most  recently,    John  has  done  the  following;  if  Ad2-SV40  ND,,    the  non-defectiv 

hybrid  of  Ad  2  and  SV40  DNA  (which,    according  to  Lewis  contains  the  ''U-ag 
gene"  of  SV40  covalently  integrated  into  the  Ad  2  chromosome)  is  denatured 
and  renatured  with  wild-type  Ad  2  DNA  one  can  observe  the  following 
heteroduplex  in  the  EM. 


•  *       I 

;«*vj  \t  -  .»  -S'^tv    u?,-l  ^   .     Q( 

There  are  two  single- stranded  (indicated  as  dashed  lines)  loops  about  16% 
in  from  one  end;  one  segment   (a)  is  longer  than  the  other.     If  SV40  Rj 
produced  linears  are  included  in  the  denaturation- renaturation  mixture  and 
the  product  is  spread  on  grids  one  sees  another  picture: 


LUOJJ  uoissiuusd  saanbai  uoponpojdaj  jaqun j  -A  juo  ysn  xniaujai  jpj  si  Adoooioqj 


220 


Dr.   M.  Stoker 
March  28,    1972 
Page  Three 


The  shorter  segment  of  the  loop  now  appears  double  stranded  and  from  it 
comes  two  single- stranded  tails.     The  length  of  the  tails  and  the  paired  loop 
is  about  equal  to  an  SV40  length.     Thus  the  R,  restriction  enzyme  makes 
a  break  near  the  U-ag  gene.     There  is  some  reason  to  believe  from  other 
Ad  2-ND  viral  DNA's  that  the  TSTA  and  T-ag  genes  are  on  the  longer  arm 
of  the  SV40  segment. 

I'd  like  very  much  to  come  to  London  for  a  bit  in  September  .    If 
you're  going  to  be  there  during  the  first  three  weeks  or  so  it  would  be 
fun  to  come  over  and  try  to  do  some  experiments.     Will  you  be  moved 
into  the  new  labs  then  or  is  that  just  about  the  time  you'll  be  moving  in? 
I'll  keep  in  touch  with  you  about  how  things  are  going  on  this  and  other 
projects.   If  you  know  of  any  appropriate  conferences  or  meetings  in 
England  or  on  the  continent  in  the  Fall,   please  let  me  know. 

So  much  for  now.   My  best  to  all  at  ICRF  and  to  Veronica.     Peter 
Beard  is  doing  very  well  and  I'm  delighted  at  his  being  here.     He's  made 
several  very  interesting  findings  that  are  proving  very  useful;  he  sends 
his  regards  to  all. 


PB/1 


'suop:»n<O 


221 
STANFORD  UNIVERSITY  MEDICAL  CENTER 

/  STANFORD.  CALIFORNIA  94305 

EPARTMENT  OF  BIOCHEMISTRY  PAUL  BERG 


Jack,  Lulu  and  Sam  IVillsoi 
Professor  of  Biochemistry 


June  19,    1972 


Dr.   Michael  Stoker 

Imperial  Cancer  Research  Fund 

P.O.    Box  123 

Lincoln's  Inn  Fields 

London,    W.  C.    2  England 

Dear  Mike, 

Another  progress  reportl    Marianne  has,   Lbelieve,   already  let 
you  know  that  the  cells  arrived  in  good  shape  and  she  will  work  them  up 
soon  to  isolate  the  DNA's  for  testing.     Thanks  for  making  that  part  of  the 
experiment  easier. 

Meanwhile  we  have  run  through  a  few  more  clones  and  repeated  the 
annealing  kinetics  with  the  older  ones.     The  results  are  quite  consistent 
and  give  the  same  result  (Table  I);    none  of  the  abortively  transformed 
BHK  have  any  trace  of  PY  DNA  I    In  Cot  #17  the  Cot1/2  w°uld  have  decreased 
by  a  2-fold  if  the  cell  DNA  had  contained  2.  5ng  of  viral  DNA;    assuming  the 
diploid  cell  genome  size  is  ^SxlO1^  daltons  of  DNA,   halving  of  the  Cotj/2 
would  have  indicated  1  viral  genome/cell.     In  Cot  #20  we  increased  the  sen 
sitivity  so  that  1.  2ng  of  viral  DNA  in  2.  5mg  of  cell  DNA  would  have  halved 
the  Cot  1/2'     As  you  can  see  except  for  clone  MT- 1  there  is  no  substantial 
homologous  sequences  in  any  of  these  abortives.     ST-1  (our  batch  of  cells) 
is  puzzling;     one  could  argue  that  it  has  about  <5l0.5  a  viral  equivalents  but 
I  don't  know  whether  to  believe  it.     "We  are  repeating  it  with  DNA  isolated 
from  the  ST-1  you  just  sent  us   (and  which  you  said  is  T-ag  negative  although 
morphologically  transformed).     We  plan  to  run  through  the  rest  of  the  abor 
tives  and  stables  we  have  to  get  more  numbers  but  then  there  is  the  question 
of  interpretation  and  what  nextl 

If  the  result  is  real  (and  I  tend  to  believe  it  at  this  point)  and  Smith  and 
Martin's  result  for  SV40  abortively  infected  cells  is  also  correct,   then  what 
is  it  trying  to  tell  us?     Is  it  that  the  steps  and  mechanism  of  transformation 
by  the  t%vo  viruses  in  these  hosts  is  different?     Conceivably  in  PY  the  inte 
gration-excision  function  (ts-a?)  is  expressed  all  the  time,   both  in  the  "free" 
and  "integrated"  state  so  that  a  DNA  molecule  is  constantly  integrating  and 
excising.     Stable  transformation  occurs  only  when  an  integrated  genome 
can't  be  excised  (even  though  excision  "enzyme"  is  present)  or  when  a  genome 
with  a  defective  ts-a  function  is  integrated  and  there  is  a  lack  of  helper  par 
ticles  to  complement  its  excision.     In  short,    wild-type  particles  can't  remain 
integrated  iznless  aberrent  insertion)  but  DNA's  with  a  defective  ts-a  function 
(as  for  examples  Mike  Fried' s  ts-a  itself  at  high  temperature)  remain  inte 
grated.     This  model  is  quite  consistent  with  Vogt  and  Summers'  and  Bill  Folk's 


r»»r»Tinnc>  'cuonTsn<v\  nnr>a(c  TO 


<Dr.   Michael  Stoker  222 

Page  Two 

• 

recent  findings  on  the  "inducibility"  of  ts-a  transformed  BHK.     In  one  sense 
BHK  may  not  be  any  different  than  mouse  cells  except  for  the  inability  to 
multiply  the  virus.     That  is,    PY  BHK  is  like  PY-3T3  and  ts-a-BHK  is  like 
ts-a-3T3  except  that  3T3  is  permissive  for  extensive  replication. 

What  about  SV40  infection  of  3T3?     Perhaps  in  this  instance  the 
integration-excision  function  is  inactivated  as  a  result  of  integration;    that 
is,    integration    shuts  off  expression  of  the  gene  controlling  the  excision 
function.     Thus  when  growth  of  the  cell  dilutes  out  existing  enzyme  and  help 
er  particles,   the  integrated  genome  becomes  "locked  in'1.     Fusion  with  per 
missive  cells  could  activate  the  excision  function  and  permit  replication  to 
occur. 

It's  quite  possible  that  there  are  clones  of  abortively  transformed 
BHK  that  do  contain  PY  DNA  but  these  might  be  rare  and  represent  only 
that  class  in  which  either  the  excision  function  was  defective  or  the  mode  of 
integration  was  abnormal;     in  either  case  the  transformed  phenotype  would 
have  to  be  "repressed". 

The  only  way  I  can  think  of  to  test  this  model  is  to  look  for  curing  of 
transformed  cells.     I  used  to  think  it  was  best  to  do  this  by  superinfecting 
PY-BHK  with  PY  (at  high  moi)  and  to  see  if  substantial  numbers  of  non-trans 
formed  clones  are  produced  (infection  should  catalyse  a  new  round  of  excision- 
integration  leading  to  a  new  stable  state,    about  5%  transformed  cells).    We 
took  a  try  at  this  but  the  attempt  was  crude,    being  done  with  only  one  strain 
of  PY-BHK;    we  have  not  done  more.    But  now  I  think  it  should  be  tried  with 
SV3T3   superinfected  with  SV40  at  multiplicities  which  cause  good  "abortive 
transformation"     I  shall  approach  Helene  Smith  about  doing  that  experiment. 

What  do  you  think?     and  what  do  you  suggest  we  do  further  with  the 
analysis  of  the  PY-BHK  abortives?     Is  it  worthwhile  making  another  batch 
of  abortives  to  test?     That  would  take  time  but  maybe  it  should  be  done  to 
nail  the  point  down.     Perhaps  one   should  use  mutagenized  PY  or  preps  en 
riched  for  defective  virus.      Conceivably  one  could  reduce  the  excision  process 
and  thereby  hope  to  produce  abortives  that  contain  viral  DNA. 

Are  you  going  to  be  in  the  States  this  summer?     At  the  Tumor  Virus 
meetings  in  Cold  Spring  Harbor  or  at  a  Gorden  Conference?     Or  is  the  only 
way  we  can  discuss  this  or  plan  something  for  me  to  come  to  London  for 
awhile?     I  turned  down  going  to  the  Brighton  Cell  Biology  meeting  (thanks  for 
suggesting  my  name  to  them  as  a  speaker)  because  the  first  two  weeks  of 
September  was  a  very  bad  time  for  me  to  be  away.     If  you're  not  going  to  be 
in  the  U.S.    do  you  believe  it  would  be  worthwhile  for  me  to  come  during  the 
second  half  of  September  for  long  enough  to  review  and  discuss  the  experiment 
and  how  to  proceed  from  here.      (Unfortunately  I  don't  think  I  can  stay  long 
enough  to  do  any  serious  experiments.  ) 

Now  for  other  news.     You  may  recall  that  I  wrote  you  about  the  Rj  restri 
tion  enzyme  making  one  specific  double- strand  scission  of  SV40  to  create  a 
unique  length  linear  molecule.      We've  used  that  molecule  now  to  locate  the 
SV40  segments  carried  by  Ad2-SV40  hybrid  ND-1,    ND-4  and  E46+  along  the 
Rj  linear. 


I  Dr.   Michael  Stoker 
Page  Three 


223 


(1)    For  example  the  Ad2  x  Ad2-ND-l  heteroduplex  looks  as  follows: 

2.  *fc«*»«  d«.W*A  fr— '  NO-t 


>cw.  •i\.'.jc-<-C   TTtv» 

and  the  heteroduplexes  formed  by  Ad2  x  Ad2-ND-l  x  RT  linears  of  SV40 
looks  like 


"foopW  A$tf*0 

-MtH*.*  R'1' 


the  short  arm  (0.1)  plus  the  short 
duplex  region  (0.  16)  and  the  long  arm 
(0.  74)  equal  one  SV40  length. 


(2)    The  Ad2  x  Ad2-ND-4  heteroduplex  is  as  follows: 


and  the  corresponding  Ad2  x  Ad2-ND-4  x  RT  linear  is 


According  to  Lewis  and  Kelly  the  AD2-ND-1  induces  U-ag  while 
Ad2-ND-4  induces  U,    TSTA  and  T-ag.     Moreover  they  say  that  little  or 
none  of  the  late  SV40  genes  are  carried  in  these  hybrids.     Therefore  it 
follows  that  the  Rj  restriction  enzyme  cleaves  in  one  of  the  "late"  genes 
of  SV40  perhaps  one  of  the  capsid  structural  genes.     One  could  draw  a 
tentative  "genetic  map"  of  SV40  (not  to  be  taken  too  seriously)  as  follows, 
The  actual  boundaries  of 


.ate       |      U 

i 

i 

TSTA              T-ag 
t                           l                       i 

Late 

)          0.  1 

0.26                   0.43               0.53 

0.  1 


rj  Xji\»Aiun  puojinns  *suo 


JDr.   Michael  Stoker 
Page  Four 

pre^-iMi.    ^Oci-^a  3«*»<i  OT  ~tlx 

TheAthree  putative  genes  are  unclear  although  the  order  seems  to  derive 
from  the  fact  that  ND- 1  induces  U  alone,   ND-2  induces  U  and  TSTA  but  not 
T  and  ND-4  induces  all  three  antigens.     It  is  interesting  that  the  segment 
assigned  to  late  genes  is  enough  to  code  for  about  70,  000  daltons  worth  of 
polypeptide  which  would  account  for  the  two  chains  40,  000  and  30,  000  that 
Renato  says  comprise  the  capsid. 

The  heteroduplexes  with  E46+  are  more  complex  but  consistent  with 
this  model.     The  difference  is  that  the  SV40  segment  integrated  in  the  E46"1" 
hybrid  has  a  deletion  of  the  Rj  restriction  site  but  the  early  genes  appear  to 
be  contiguous. 

The  most  recent  finding  which  excites  us  is  that  Rj  makes  a  staggered 
break,    i.  e.  ,  I  in  which  the  number  of  bases  between 


breaks  is  of  the  order  of  6i2  (probably  six).     Thus  the  linears  can  be  circu 
larized  at  low  concentration  at  3-5°C  and  can  be  covalently  sealed  with  DNA 
ligase  to  regenerate  completely  infectious  and  full  length  SV40  molecules 
(actually  the  linears  are  one-tenth  as  infectious  as  wild-type  probably  because 
the  cell  itself  can'blose  the  ring").  It  seems  very  likely  that  the  site  at  which 
the  enzyme  cleaves  is  identical  in  all  DNA's   (it  occurs  on  the  average  once 
per  4,  000  bases  in  a  random  sequence)  and  very  likely  is  summetrical. 

i  -J 

-  i^A       B        C        C'      B1      A'  - 

-  A1       B'      C'      C        B 


We  can  show  experimentally,   that  any  two  DNA  molecules  having  ends  pro 
duced  by  RT  endonuclease  can  be  covalently  joined.     In  other  words  the  ability 
to  construct  molecular  hybrids  is  enormously  extended.     We  have  now  to  find 
out  how  to  deal  with  this  intelligently. 

How  is  the  work  going  on  the  serum  factors?     Is  it  possible  to  assign 
specific  cellular  functions  to  specific  serum  proteins  yet?     You  must  be  just 
about  getting  ready  to  move  to  the  new  quarters.     Good  Luck.     I  envy  Art 
Pardee  his  next  year. 

Well  so  much  for  now.     I  look  forward  to  hearing  from  you  about  PY 
abortives.     As  we  get  more  data  I'll  keep  you  in  touch.     I'd  appreciate  it  if 
you  could  pass   some  of  this  scientific  information  along  to  Bill  Folk  and  any 
body  else  you  care  to  so  as  to  save  me  the  time  of  writing  it  to  him. 

With  best  regards  to  all  at  ICRF  and  to  Veronica. 


Sincerely  yours, 


~.  ,£  ) 

PB:af 


rmnirfai  t3inm  j   'Xnio  a»n  mroEJiai  jm  <n 


for.   Michael  Stoker 
T>age  Five 

i 

P.S.        I  wonder  if  I  can  make  a  request  of  you.     So  far  we've  been  using 
one  of  our  PY  virus  stocks  to  prepare  the  p-^2  DMA  for  annealing.     That's 
not  the  same  one  we  used  to  do  the  infections  of  BHK.     I  don't  believe  that's 
serious  but  perhaps  we  should  make  some  DNA  from  your  stock  to  be  sure. 
More  importantly  our  virus  stock  must  surely  contain  defective  particles 
containing  insertions  of  host  DNA.     This  slightly  complicates  the  kinetics  of 
annealing  (because  the  host  sequences  probably  don't  anneal  with  the  same 
kinetics  as  the  PY  sequences).     If  your  stock  is  plaque  purified  or  if  you 
have  a  plaque  purified  stock  from  which  you  could  let  us  have  enough  to  make 
a  batch  of  DNA  (from  infected  mouse  kidney  cells)  that  would  be  very  help 
ful.     Otherwise  we  shall  have  to  take  the  time  to  make  such  an  isolate  and 
that  would  be  time  consuming. 


puojirns  'suoiKwn°D  (toads  J°  nwumwfaQ 


226 


TABLE  I 


Cell  DNA's  tested 


1 


Cot  #17  Cot  #20' 

Normalized  Cot.  ,., 


Salmon  Sperm 

BHK 

SA-2 

SA-10 

MA-4 
MA-6 
MA- 8 
ST-1 
MT-1 


2.  1  x  10 


-3 


2.  1  x  10 


-3 


2.0  x  10 


-3 


1.9  x  10 
1.6  x  10 


-3 


-3 


0.6  x  10 


-3 


1.3  x  10' 

1.3  x  10 
1.9  x  10 

1.4  x  10 
1.3  x  10 

1.3  x  10 

1.4  x  10 
0.  9  x  10 


-3 


-3 


-3 


-3 


-3 


-3 


1.  Cell  DNA's  were  added  to  the  annealing  mixture  at  a  concentration 
of  50A26Q  (2«5mg/xnl);    it  was  sheared  to  average  single-strand 
chain   length  of  400-500  bases. 

2.  In  Cot  #17  the  32P-PY  DNA  was  at  a  concentration  of  5  x  10"5A260 
(2.5ng/ml);    average  chain  length  of  DNA  was  400-500  bases. 

3.  In  Cot  #20  the  32P-PY  DNA  was  at  a  concentration  of  2.  5  x  10~5A260 
(1.  2ng/ml);    average  chain  length  same  as  in  Cot  #17. 


an  UJQJI  uoissiuuad  sannbai  uoncmpoidai  JTUUTI  4  'Xruo  ?sn  xniaiaiai  jpi  si 


Photocopy  is  for  reference  use  only.  Further  reproduction  requires  permission  from  the 
Department  of  Special  Collections,  Stanford  University  Libraries 


227 


March  27,    1973 

Dr.  Norman  K.  Wet selli 
Department  of  Biology 
Stanford  University 

Dear  Norm, 

-.  I  want  to  nominate  Miss  Janet  Mertz.for  this  year1* 

Francis  Lou  Kail  man  Memorial  Award.    It's  zny  view  that 
Janet  i*  one  of  the  beet  graduate  students  presently  in  our 
department;    in  fact*  I  would  rank  her  amongst  the  top  five 
students  we  have  ever  had  in  Biochemistry.    She  is  one  of 
the  hardest  working,  brightest  and  most  creative  young 
people  I  have  ever  worked  with.     Moreover,   she  has  that 
rarest  of  talents:    things  get  done  no  matter  the  difficulties 
encountered.    I  predict  a  very  bright  and  productive  scienti 
fic  career  for  her  future. 

Janet  came  to  our  Department  in  September  of  1970 
with  a  very  distinguished  undergraduate  record  from  MIT; 
she  completed  a  double  major  in  electrical  engineering  and 
biological  sciences  in  three  years  with*  as  I  recall,  almost 
a  straight  A  record.    The  enthusiasm  with  which  we  accepted 
her  has  not  diminished  one  iota. 

From  the  day  she  began  research  her  progress  has 
been  most  impressive.    Her  project  was  to  examine  the 
possibility  of  isolating  deletion  mutants  of  SV40  virus  and 
with  these  to  locate  the  genes  coding  for  the  different  viral 
functions.    With  almost  breathtaking  speed  she  mastered  the 
available  literature  and  set  about  acquiring  the  cell  culture 
skills  needed  to  begin  her  experiments.    I  was  astonished  at 
how  rapidly  she  mastered  these  techniques  and  how  frequently 
•he  came  up  with  improvements  in  our  standard  procedures. 
Now,  she  is  one  of  the  "experts"  of  our  group  and  I  would 
count  heavily  on  her  abilities  to  train  newcomers  to  the  lab. 


,~      .* 


use  viuy.  r-ujmcr  reproduction  requires  permission  from  the 
Department  of  Special  Collections,  Stanford  University  Libraries 


228 


Dr.  Norman  Wes cells 
Page  Two 


Janet  has  virtually  completed  three  publishable  and 
quite  significant  researches. 

1)  Together,  with  Ron  Davis,   she  discovered  that  the 

RI  restriction  endonuclease  catalyzed  cleavage  of  DNA  generates 
identical  and  cohesive  ends;    this  makes  it  possible  to  reseal 
the  ends  of  a  circular  molecule  cleaved  by  this  enzyme  or.  more 
significantly,  to  join  any  twoJjN A  molecules  or  fragments  having 
such  termini  (see'enclosed'reprint)^  (Yanofsky  is  presently 
using  this  approach  for  joining  the  tryptophan  represser  gene 
to  a  p  Las  mid  form  of  X  DNA  so  that  the  number  of  copies  of  the 
represser  gene  can  be  increased  in  cells  made  to  carry  that 
plasmid.  )    Her  presentation  of  this  work  at  the  Tumor  Virus 
Meeting  at  Cold  Spring  Harbor  created  quite  a  stir. 

2)  Together  with  one  of  Dale  Kaiser's  postdoctoral 
fellows,   Doug  Berg,  whe  prepared  and  isolated  what  has  turned 

/     out  to  be  an  extremely  useful  new  type  of  genetic  structure: 
Xdvgal.     In  this  structure  the  gal  operon  of  E.  coli  has  been 
fused  into  a  small  DNA  plasmid  containing  about  7%  of  the  X  phage 
genome.     These  molecules  can  be  propagated  in  E.  coli  cells  as 
episomes  (about  50.100  copies  per  cell).     Janet  modified  and 
improved  an  assay  system  for  introducing  the  pureXXdvgal  DNA 
into  virgin  cells  to  reestablish  the  episomal  state.     This  system, 
therefore,  provides  a  way  of  introducing  new  genes  into  E.  coli; 
by  covalently  joining  any  piece  of  DNA  to  the  Xdvgal  DNA,  it 
can  be  introduced  and  stabilised  in  the  recipient  bacterial  cell. 
This  ability  has  paved  the  way  for  Dave  Hogness*  group  to 
attempt  to  clone  Drosophilia  DNA  segments. 

3)  Janet  is  now  well  along  in  the  main  part  of  her  thesis 
research:    The  isolation  and  characterization  of  deletion  mutants 
of  SV40.     She  has  already  prepared  and  characterized  stocks 
with  a  variety  of  deletion  and  substitutions  at  various  locations 
in  the  SV40  genome.     In  doing  this  she  has  become  an  expert  and 
sophisticated  electron  micros copiat  and  with  this  technique  has 
carried  out  an  elegant  analysis  of  the  heteroduplexes  formed 
from  deletion* substitution  mutant  DNA'*  with  wild-type  molecules. 
Her  next  immediate  goal  is  to  clone  these  and  to  characterize  the 
specific  genetic  defect  each  type  of  deletion  produces. 


Photocopy  is  for  reference  use  only.  Further  reproduction  requires  permission  from  the 
Department  of  Special  Collections,  Stanford  University  Libraries 


229 


Dr.  Norman  Wes  sells 
Page  Three 


Let  me  say  in  finishing  that  Janet  is  one  of  the  most 
eager  student  teachers  we've  had.    As  a  first  year  student 
she  volunteered  (she  was  the  only  one  to  do  that)  to  supervise 
a  section  of  almost  15  students  in  our  Biochemistry  201  course. 
The  following  year  she  participated  in  literature  discussions 
with  another  group  of  Biochemistry  students.     She  generally 
gives  some  of  the  best  seminars  in  courses  or  in  Departmental 
meetings.    Her  performance  as  well  as  her  research  has 
already  attracted  considerable  attention  at  several  National 
Meetings. 

In  summary,  then,  let  me  say  that  I  know  of  no  student, 
let  alone  any  woman  student,  that  I  feel  more  confident  about 
in  nominating  for  this  recognition.     She  has  certainly  earned  it. 

Sincerely  yours, 


Paul  Berg 

Professor  and  Chairman 


PB:af 
Enc. 


Photocopy  is^fiorreference  use  only.  Further  reproduction  requires  permission  from  the 
Department  of  Special  Collections,  Stanford  University  Libraries 


230 


September  16.    1975 

Dr.   William  J.   Rutter 

Department  of  Biochemistry  and  Biophysics 

University  of  California 

San  Francisco,  California  94143 

Dear  BUI. 

I'm  delighted  to  be  able  to  support  your  recommendation 
that  Hejrb^Bovjsr  be  promoted  from  Associate  Professor  to  full 

ProfeTTort"* 

Herb's  contributions  over  the  past  five  years  on  the  mechanisms 
of  restriction  and  modification  of  DNA  by  microbial  and  phage  enzymes 
have  been  outstanding.     His  work  combined  elegant  genetic  experiments 
with  enzyme  studies  to  sort  out  the  kind  of  relationships  which  charac 
terize  the  restriction-modification  specificities. 

About  two  years  ago  his  work  took  another  and,   I  believe,   more 
original  and  germinal  line.     This  was  to  isolate  purified  restriction 
and  modification  enzymes,   to  characterize  the  nature  of  their  cleavages 
and  modifications  chemically.     During  these  studies  two  enzymes  (EcoRI 
and  EcoRII)  which  had  been  discovered  and  purified  in  Boyer's  lab  were 
found  to  have  remarkable  properties:    they  cleave  both  strands  of  DNA's 
lacking  the  appropriate  modifications  at  specific  p%lrmdromic  sequences 
so  as  to  create  cohesive  ends.     Not  only  do  these  enzymes  serve  as 
site-specific  nucleases,   making  possible  nucleotide  sequencing  of  the 
fragments  but  the  existence  of  cohesive  ends  makes  possible  in  vitro 
recombination  between  different  DNA  molecules  having  the  same  enzyme- 
generated  cohesive  ends.     This  may  well  revolutionize  molecular 
genetics  as  indicated  by  the  astounding  experiment  by  Boyer  and  his 
colleagues  in  which  they  succeeded,  in^synthesizing  hybrid  DNA  molecules 
containing  the  16S,    28S  or  both  rDNA'.'.ot  kenopus^ciavaUntly  inserted  into  -*' 
tive"  circular  R  factor  DNA  and  to  propagate' these  molecules  in  growing 
E.   coli  as  aa  episomal  elements     The  implications  of  this  accomplishment 
are  enormous  and  Herb  is  actively  pursuing  many  of  these  new  leads. 

Boyer's  work  has  had  great  influence  on  the  newer  recognition 
that  restriction  enzymes  provide  us  with  a  very  powerful  methodology  for 
the  analysis  of  the  structure  and  expression  of  DNA  chromosomes  of 
viruses,  bacteria  and  higher  cells.     I  believe  Herb  is  one  of  the  world's 
leaders  in  the  field  of  restriction  modification  and  particularly  in  its 
application  to  trther  broader  problems  such  as  gene  expression.chromoson 


Photocopy  is  for  reference  us^^^i.  Fustbei  reproduction  requires  permission  from  the 
Department  of  Special  €0flecrions,  Stanford  University  Libraries 


231 


Dr.   William  J.  Rutter 
Page  Two 


structure.     His  work  in  the  construction  and  study  of  recombinant 
DNA  molecules  will  certainly  be  at  the  forefront  of  this  area  of  biology 
for  a  long  time  to  come. 

You  may  recall  that  I  nominated  Herb  for  the  1975  Pfizer  Award 
in  Enzyme  Chemistry.     That  is  one  measure  of  the  high  regard  I  have 
for  his  contributions. 

I  should  also  add  that  my  scientific  contacts  with  Herb  have 
always  been  most  cordial  and  helpful.     Time  and  time  again  he  has  most 
generously  given  us  enzyme  preparations,  bacterial  strains  and  unpub 
lished  data.     In  today's  world  of  "cutthroat"  competition,   I  regard  his 
behavior  as  being  in  the  best  traditions  of  open  science. 

With  best  regards. 
Sincerely, 


Paul  Berg 

Professor  of  Biochemistry 

PBraf 


*  232 


Testimony  by  Paul  Berg 

Subcommittee  on  Science,  Technology  and  Space 
November  2,  1977 

Senator  Stevenson,  I  am  grateful  for  your  invitation  to 
participate  in  this  committee's  discussion  of  the  current  status 
of  recombinant  DNA  activities.   I  particularly  value  the  oppor 
tunity  to  present  my  views  on  the  fundamental  and  practical  issues 
that  have  been  raised  in  the  public  debate  on  recombinant  DNA 
methods. 

To  begin,  let  me  introduce  myself.   My  name  is  Paul  Berg 
and  I  am  Willson  Professor  of  Biochemistry  at  Stanford  University 
School  of  Medicine.   When  I'm  not  distracted  by  recombinant  DNA 
matters  I  conduct  research  and  teach  biochemistry  and  molecular 
•biology.   My  particular  specialties  are  molecular  genetics  and 
viral  carcinogenesis,  both  of  which  have  become  increasingly  amen 
able  to  and  dependent  upon  the  use  of  recombinant  DNA  methods. 
I  have  neither  a  direct  nor  indirect  association  with  any  commer 
cial  enterprise  engaged  in,  or  contemplating,  research  or  manu 
facture  using  recombinant  DNA  methods. 

I  am  also  not  a  newcomer  to  the  recombinant  DNA  controversy. 
A  moment  will  suffice  to  summarize  the  extent  of  my  involvement. 
My  laboratory  was  amongst  the  first  to  construct,  outside  of  a 
living  cell,  a  hybrid  or  recombinant  DNA  molecule;  hence,  I  was 
one  of  the  earliest  practitioners  of  recombinant  DNA  research. 
Because  several  friends  and  colleagues  expressed  concern  about 
the  ramifications  of  my  experiments  I  became  an  early  partici 
pant  in  discussions  of  their  potential  risks.   Subsequently,  my 
involvement  with  these  concerns  grew  by  being  chairman  of  a  commit 
tee  that  warned  the  National  Academy  of  Sciences  about  possible 
risks  that  might  result  from  the  indiscriminate  use  of  recombinant 
DNA  methods.   I  also  served  as  chairman  of  the  committee  that 
convened  and  presided  over  the  Asilomar  Conference  on  Recombinant 
DNA  Molecules;  the  report  of  those  proceedings  to  the  National 
Institutes  of  Health  made  specific  and  novel  recommendations  for 


233 
-2- 

scientific  and  administrative  procedures  that  could  ensure  safe 
conduct  of  this  line  of  research.   Although  not  one  of  the  archi 
tects  of  the  NIH  Guidelines,  I  was  consulted  at  various  times 
during  their  formulation  and  prior  to  their  release  in  July,  1976. 

A  relevant  question  with  which  to  begin  is  why  are  biologists 
throughout  the  world  so  excited  by  the  recombinant  DNA  methodology. 
Is  it,  as  some  have  charged,  just  fun  and  games,  the  chance  to 
enhance  one's  career  or  ambitions  or  is  it  to  advance  genetic  mani 
pulation  of  humans  for  nefarious  purposes?   I  doubt  that  any  of 
these  selfish  reasons 'motivate  more  than  a  small  fraction  of  the  interna 
tional  scientific  conrnunity.  Rather,  the  overwhelming  body  of  scientists  view 
the  recombinant  DNA  methodology  as  an  extraordinary  opportunity 
to  solve  important  biological  problems;  the  knowledge  gained 
will  illuminate  our  biologic  nature  and  heritage;and  very  likely, 
help  to  alleviate  the  tragedies  of  human  disease,  starvation  and 
the  pollution  of  our  environment.   What  are  the  opportunities,  and 
important  biological  problems  that  recombinant  DNA  research  can 
help  to  solve?   Basically  there,  are  three  answers: 

1)  The  recombinant  DNA  methodology  permits  the  isolation  of 
single  or  groups  of  genes  in  high  purity  and  virtually  unlimited 
quantities  from  almost  any  living  organism.   Except  in  special 
cases  this  can  not  be  accomplished  by  any  other  presently  available 
method.   Coupled  with  another  new  procedure,  that  is  virtually 
child's  play,  the  basic  chemical  structure  of  these  isolated  genes 
can  be  readily  solved.   These  two  techniques  can  tell  us  a  great 
deal  about  the  molecular  structure  and  organization  of  the  complex 
chromosomes  of  higher  plants,  animals  and  man.   I  described  how 
recombinant  DNA  methods  were  uniquely  suited  for  the  task  of  recon 
structing  complex  chromosomes  during  my  presentation  to  the  National 
Academy  of  Sciences  Forum  on  Recombinant  DNA  Research  last  March. 
These  are  not  idle  speculations.   They  are  realistic  estimates 
drawn  from  the  impressive  achievements  so  far.   There  have 

also  been  problems  and  several  surprises;  each  of  the  surprises 
introduces  unexpected  subtleties  and  makes  more  fascinating 
and  urgent  that  we  get  on  with  their  solution. 

2)  The  ability  to  join  together  different  DNA  molecules  per- 


234 
-3- 


in  simple  as  well  as  complex  chromosomes.   Together  with  classical 
methods  for  creating  hybrid  cells  and  organisms,  one  can  envision 
more  sophisticated  analyses  of  the  mechanism  of  gene  and  chromo 
some  function.   Understanding  differentiation,  the  process  where 
by  embryonic  cells   containing  the  identical  complement  of  genes 
and  chromosomes,  gives  rise  to  the  myriad  cells  and  organs  of  the 
organism,  is  a  worthwhile  and  realizable  goal.   It  is  difficult 
for  me  to  see  how  that  knowledge  will  not  have  ramifications  for 
the  treatment  and  possibly  prevention  of  certain  birth  defects 
and  other  developmental  disorders. 

3}   The  ability  to  isolate  pure  genes  puts  us  at  the  thresh 
old  of  new  forms  of  medicine,  industry  and  agriculture.   Tailor- 
made  organisms  produced  by  recombinant  DNA  methods  could  provide 
valuable  diagnostic  reagents,  probes  for  studying  the  operational 
status  and  efficiency  of  gene  expression  in  health  and  disease, 
vaccines  to  immunize  individuals  and  animals  against  the  ravages 
of  certain  bacterial  and  viral  infections  and,  possibly,  even 
cancer;  and,  finally,  there  is  extraordinary  progress  towards  the 
construction  of  organisms  that  make  therapeutically  useful  pro 
tein  hormones;  the  isolation  of  the  insulin  gene  is  a  promising 
start;  the  bacterial  production  of  somatostatin,  a  hormone  produced 
by  the  brain  is  even  more  astonishing.  A  joint  effort  between  research  groups  at 
the  University  of  California  Medical  Center,  San  Francisco,  the 
City  of  Hope  in  Los  Angeles  and  the  Salk  Institute  in  San  Diego 
has  resulted  in  the  production  of  about  5  mg  of  somatostatin; 
only  100  gms  of  E.coli ,  grown  in  about  2  gallons  of  culture  was 
needed.   Bear  in  mind  that  it  took  nearly  half  a  million  sheep 
brains  to  yield  5  mg  of  somatostatin  in  the  researches  for  which 
Drs.  Guillemin  and  Schalley  received  this  year's  Nobel  Prize  in  Medicine. 
Equally  significant  is  the  ingenious  and  elegant  way  in  which 
it  was  accomplished;  chemical  synthesis  of  the  gene  and  pro 
duction  of  a  modified  form  of  the  hormone  so  that  chemical 
processing  outside  the  organism  is  necessary  to  liberate  the. hormone 
This  approach  provides  a  novel  alternative  to  the  previously  planned 
procedures  for  producing  many  such  products. 


235 
-4- 


In  this  brief  statement  I  can  only  mention  but  not 
amplify,  some  of  the  important  advances  that  are  being  made  by 
recombinant  DNA  techniques.   If  you  like  I  could  expand  on  some 
of  them  in  the  subsequent  discussion.   In  short,  I  sense  a  mounting 
wave  of  accomplishment  and  progess  that  give  lie  to  the  charge  that 
the  benefits  of  reccmbinant  DNA  research  are  only  speculative 
and  ephemeral,  and  that  only  the  dangers  are  real. 

Before  considering  the  question  of  risks  I  want  to  say 
.a  few  words  about  genetic  engineering,  -  the  directed  modi 
fication  or  even  construction  of  new  genetic  constitutions  for 
animals,  plants  and  man.   Partly  because  of  the  exaggerated 
and  misleading  claims  by  the  popular  press  and  some  scientists 
and  laymen  as  well,  this  term  has  evoked  as  much  alarm  as  excite 
ment.   I  would  guess,  that  deep  down  it  is  what  troubles  some 
people  most.   But  man  has  been  involved  actively  in  genetic  engi 
neering  ever  since  he  came  down  from  the  trees,  planted  maize  and 
domesticated  animals.   The  animals  and  plants  that  provide  our 
food,  the  microorganisms  that  make  our  bread,  beer  and  wine, 
the  organisms  that  make  our  antibiotics  and  purify  our  sewage, 
are  all  subject  to  our  genetic  counseling.   We  have  carried  out 
wars  of  genocide  against  polio  virus,  small  pox  and  plague  and 
are  much  the  better  for  it.   Recall  that  for  the  worst  holo 
caust  in  history  Hitler  did  not  need  science  and  technology; 
ovens  and  gas  chambers  did  the  job.   Malnutrition,  poor  and  inad 
equate  nutrition  warps  the  minds  and  bodies  of  hundreds  of 
millions  of  infants  and  children  throughout  the  world   and  our 
personalities  and  behavior  are  manipulated  and  profoundly  in 
fluenced  by  the  printed  page  and  television.   Genetic  manipulation, 
then,  is  not,  itself,  good  or  bad;  we  need  to  distinguish  between 
the  acquisition  of  knowledge  and  the  applications  of  that 
knowledge  and  know  how  to  achieve  both  wisely.   Human  gene 
tic  engineering  is  a  concept  worth ' examining  in  rational 
ways.   It  is  not  at  all  clear  that  it  is  feasible,  nor  when 
it  will  be,  if  at  all.    There  are  many  difficult  and 
contentious  scientific, ethical  and  moral  questions  to  be  examined 
and  at  many  stages  there  will  be  opportunities  by  all  segments 
of  our  society  to  have  their sav.   But  preventing  or  slowing  down 


236 
-5- 

basic  genetic  research  now,  seems  ill-suited  to  dealing  with 
that  question. 

Nov;  let  me  turn  to  the  matter  of  risks.   Three  years  ago 
I  expressed  concern  about  the  use  of  recombinant  DNA  techniques. 
There  was  no  evidence  that  such  experiments  were  hazardous,  only 
conjecture;  but  we  wanted  assurance  that  these  novel  experiments 
would  be  safe.   More  than  three  years  later,  after  considerable 
discussion  by  experts  in  this  country  and  abroad  and  the  analyses 
of  past  experiences  and  new  findings,  I  and  others  have  changed 
bur  assessment  of  the  risks.   I  now  believe  that  the  possibility  • 
that  experimental  organisms  will  be  hazardous  or  released  is 
exceedingly  small. 

Where  it  has  been  examined,  organisms  modified  by  recombinant 
DNA  methods  are  at  a  disadvantage  in  competing  with  their  paren 
tal  or  wild  organisms.   Moreover,  certain  constructed  DNA  molecules, 
hitherto  believed  to  be  novel,  can  arise  in  nature  by  reactions 
akin  to  those  used  in  the  laboratory.   There  is  also  the  virtually 
unanimous  agreement  of  experts  in  infectious  disease  and  epidemiolo 
gy  that  strain  K12 ,  the  enfeebled  laboratory  variant  of  E.  coli 
widely  used  for  recombinant  DNA  experiments,  is  unable  to  colonize 
normal  hunan  or  animal  intestinal  tracts.    Based  on  recent  experi 
ments  and  existing  data,  these  experts  also  concluded  that  there 
is  little  or  no  likelihood  that  strain  K12  can  be  transformed 
into  an  infectious  or  pathogenic  organism  or  even  into  a  human 
intestinal  inhabitant  by  a  bit  of  foreign  DNA.   This  view  has 
been  echoed  by  Rene  Dubos  one  of  our  most  eminent  biologists,  an 
authority  in  infectious  diseases  and  an  ardent  environmentalist. 
He  concluded  that  "I  doubt  that  gene  recombination  in  the  labora 
tory  will  create  microbes  more  virulent  than  those  endlessley 
created  by  natural  processes".   Moreover,  the  introduction  of 
genetically  enfeebled  derivatives  of  strain  K12  and  vectors  that 
are  not  readily  transmissable  to  other  bacteria,  provide  a  fur 
ther  measure  of  safety.   Hence,  our  initial  concern  that  novel  and 
laboratory-created  recombinant  DNA  molecules  could  become  widely 
disseminated  to  man,  animals  and  the  ecosystem  is  not  supported 
by  the  available  data.  / 

Enacting  legislation  to  govern  the  content  and  methods  of     >/ 


-6- 


In  my  view  legislation  of  the  type  that  has  so  far  been  pro 
posed  would  inhibit  basic  research  on  important  biological  and 
medical  problems.   The  rules,  procedures,  and  penalties  are  pre 
dicated  on  assumptions  that  will  surely  change,  thereby  making 
it  difficult  and  cumbersome  to  adjust  to  the  changing  information, 
ideas  and  opportunities.   I  believe  that  legislation  could  stul 
tify  the  creativity  and  initiative  that  has  characterized  the 
development  of  the  recombinant  DNA  technique;  it  could  also 
discourage  and  disillusion  young  scientists  from  entering  this 
field.   I  believe  that  the  present  U.S.  NIH   guidelines,  as  well 
as  analogous  codes  of  practice  in  other  countries,  afford  the 
security  to  meet  the  perceived  risks.   Many  scientists  believe 
the  guidelines  are  too  restrictive  and  that  most  of  the  pro 
scriptions  cannot  be  justified  by  any  scientific  information  we 
now  possess.   But  in  spite  of  their  reservations,  scientists  and 
their  institutions  have  accepted  the  guidelines  as  an  interim  so 
lution  to  the  anxieties  that  remain.   The  acceptance  of  that  view 
is  a  responsible  action  based  on  careful  weighing   of  the  alterna 
tives  and  rejects  irrational  fears  as  a  basis  for  decision. 

As  I  see  it,  most  of  us  are  seeking  the  same  objective:  To    v 
reap  the  benefits,  basic  knowledge  and  practical  advances  from 
recombinant  DNA  research  with  a  minimun  of  risk  to  our  world. 
.Members  of  the  academic  research  community  are  now  the  principal 
practitioners  of  recombinant  DNA  research  in  this  country.   Since 
most  of  their  research  is  funded  by  government  agencies, it  is  being 
done  in  compliance  with  the  procedures  and  administrative  mecha 
nisms  embodied  in  the  NIH  Guidelines.   The  sanctions  and  con 
sequences  are  severe  and,  therefore,  a  strong  deterrent  to  non- 
compliance.   A  question  frequently  put  is  •-  what  about  recombinant 
DNA  activities  that  are  not  under  the  Guidelines'  jurisdiction? 
But  surely  there  are  existing  mechanisms  that  guard  the  public 
against  known  hazards  of  pathogenic  agents.   Are  there  not  exist 
ing  statutes  that  could  deal  with  these  .  hypothetical  risks  as  they 
do  now  with    real  and  documented  hazards?   If  not,  we  could  consid« 
establishing   a  parallel  set  of  procedures  and  practices, agreed 
to  by  representatives  of  the  private  sector  and  monitored  by  the 
Department  of  Commerce,  to  cmidp  i  nrhist-r ial  r^sparch.  develooment 


238 


and  production  activities  using  recombinant  DNA  methods? 
Industry's  concerns  in  this  area  are  unique  to  them;  and  the 
academic  research  community's  concerns  are  foreign  to  the  world 
of  commerce.   Does  it  make  sense,  then,  to  have  both  types  of 
activity  operate  by  an  identical  set  of  rules  and  procedures 
and  subjected  to  constraints  that  are  inappropriate  to  each? 
I  suspect  that  just  as  the  consortium  .  of  scientists,  the  public 
and  the  Department  of  HEW  arrived  at  acceptable  codes  of  prac 
tice,  a  similar  coalition  of  the  industrial  sector,  the  public 
and  the  Department  of  Commerce  could  develop   an  equally  accept 
able  set  of  guidelines  for  their  activities. 

Let  me  end  by  saying  that  I  am  particularly  concerned  by  the 
growing  efforts  and  influence  of  the  anti-science  forces. .   This 
is  apparent  in  the  increasing  pressures  to  suppress  scientists' 
explorations  for  fear  of  what  their  discoveries  will  uncover  or 
produce.   Decisions  and  agreements  about  what  is  desirable, 
acceptable  and  safe  to  know  are  nearly  impossible  to  obtain  at 
each  level  of  social  organization.   Deeply  held  and  conflicting 
sociopolitical  ideals  challenge  the  traditional  views  of  what 
science  is  for  and  how  it  should  be  done.   As  these  forces  gain 
momentum,  there  are  increasing  attempts  to  restrict  scientific 
research. 

Society  desperately  requires  effective  mechanisms  for  anti 
cipating  and  evaluating  the  impact  of  scientific  and  technologic 
breakthroughs.   In  the  recombinant  DNA  matter  scientists  demons 
trated  that  they  could  provide  the  early  warning  system  for  alerting 
society  to  the  potential  benefits  and  risks  of  their  discoveries; 
accusations  of  self-interest,  arrogance  or  even  malevolence  do 
little  to  encourage  further  efforts  of  that  kind.   We  may  already 
have  squelched  the  concerned  scientist  of  tomorrow.   Governing 
bodies,  everywhere,  must  seek  better  ways  to  encourage  scientists' 
participation  and  the  means  to  channel  their  input  into  the  determi 
nation  of  policy. 

Perhaps,  these  poetic  words  of  Aristotle  can  guide  us,   scientists  and 
politicians,  in  our  search  for  wisdom  in  these  matters. 


-8-  239 


He  wrote: 


"The  search  for  truth  is  in  one  way  hard  and 
in  another  easy.   For  it  is  evident  that  no 
one  can  master  it  fully  nor  miss  it  wholly. 
But  each  adds  a  little  to  our  knowledge,  and 
from  all  the  facts  assembled  there  arises  a 
certain  grandeur." 


Thank  you, 


2 

,0 

— 
>, 

I 

> 

'c 


O 


o 

U 


_,  .  240 

Photocopy  is  lor  reference  use  only.  Further  reproduction  requires  permission  from  the 

Department  oETp^flOIBII^filMi^SEftfidaiIrM6fi^^bi*i^TER 

STANFORD,  CALIFORNIA  94305 

TMENT  OF  BIOCHEMISTRY  PAUL  BERG 

Willson  Professor  of  Biochemistry 

March  5,   1979 


Dr.  Richard  A.  Rifkind 

Cancer  Center/Institute  of  Cancer  Research 

College  of  Physicians  &  Surgeons  of 

Columbia  University 
701  West  168th  Street 
New  York,  New  York  94305 

Dear  Dr.  Rifkind, 

I  really  wish  I  had  the  talent  to  pick  up  a  dictaphone  and  provide  a 
scholarly,  comprehensive  summation  of  the  state  of  and  opportunities  in 
molecular  genetic  research;  but  I  don't.   I  also  wish  I  had  the  leisure 
to  organize  various  thoughts  and  opinions  on  these  subjects  in  a  letter, 
but,  alas,  that  too  is  lacking.   In  fact  other  pressing  obligations  that 
can  not  be  put  off  have  precedence  on  my  time  and  energy,  thereby  necessi 
tating  a  brief  reply. 

In  my  view  the  most  promising  opportunities  in  molecular  genetics 
research  for  now  and  the  coming  decade  will  be  in  the  area  of  organization, 
replication,  expression  and  regulation  of  mammalian  (human)  and  other 
eukaryote  genomes.   The  emergence  of  restriction  endonuclease  and  molecular 
cloning  techniques,  rapid  DNA  sequencing  and  the  ability  to  prepare  mono 
clonal  antibodies  make  feasible  experimental  approaches  to  many  problems, 
that  were  hitherto  impossible.   I  believe  it  will  be  possible  to  reconstruct, 
in  molecular  detail,  the  gene  organization  of  specific  loci  (e.g.  the  loci 
governing  the  expression  of  the  human  hemoglobins,  the  innuno globulins  and 
HL-A  regions  are  in  the  offing)  as  well  as  extended  chromosomal  regions. 
I  also  believe  we  may  be  able  to  identify  the  genetic  signals  and  mechanisms 
that  govern  differential  gene  expression  (e.g.  hormone  control,  and  other 
homeostatic  mechanisms)  and  possibly  to  define  the  general  features  of 
developmental  programs.   Clearly  if  that  comes  to  pass  the  impact  on  our 
understanding  of  the  underlying  mechanisms  of  many  pathologies  will  be 
profound.   The  progress  I  foresee  will  enable  us  to  reap  the  rewards  of 
the  basic  molecular  biology  advances  of  the  last  three  decades. 

Without  being  exhaustive  I  would  identify  the  following  individuals 
as  people  capable  of  making  giant  strides  in  that  direction:   David  Hogness, 
Donald  Brown,  Richard  Axel,  Philip  Leder,  Tom  Maniatis,  Phillip  Sharp, 
Charles  Weissmann,  Pierre  Chambon,  Richard  Flavel,  David  Botstein, 
Ronald  Davis,  Gerald  Fink.   Undoutedly  there  are  others  but  these  will 
give  you  an  idea  of  the  type  of  people  I  have  in  mind. 

Good  Luck  in  your  venture, 

Sincerely, 


241 


APPENDIX   C 


The  1980  Nobel  Prize  in  Chemistry 


Three  molecular  biologists  win  the  prize  for  discoveries 
that  can  be  used  to  study  gene  structure  and  control 


The  current  Nobel  Prize  in  Chemistry 
spotlights  contributions  to  the  methodo 
logical  revolution  that  is  allowing  re 
searchers  to  examine  the  structure  and 
control  of  genes  of  higher  organisms  in  a 
dc'.'.''  previously  nnimngined.  Half  of  the 
prize  was  awarded  to  Paul  Beig  of  Stan 
ford  University;  the  other  half  was 
.  awarded  jointly  to  Frederick  Sanger  of 
•  Cambridge  University  and  Walter  Gil 
bert  of  Harvard.  This  is  Sanger's  second 
Nobel  Prize. 

Berg  is  cited  for  "his  fundamental 
studies  of  the  biochemistry  of  nucleic 
acids,  with  particular  regard  to  recombi- 
nj.  :  DNA."  According  to  a  press  re 
lease  from  the  Swedish  Royal  Academy. 
"Berg  was  the  first  investigator  to  con 
struct  a  recombinant  DNA  molecule. 
i.e..  a  molecule  containing  parts  of  DNA 
from  different  species.  His  pioneering 
experiment  has  resulted  in  the  develop 
ment  of  a  new  technology,  often  called 
genetic  engineering."  Berg  does  not 
kr.iiw  whether  the  Nobel  committee  had 
a  ;-.ir:icu!dr  experiment  in  mind  but.  he 
sav  -,.  "1  would  like  to  think  it  [the  prize] 
was  for  a  body  of  work  and  not  for  a 
single  experiment. "  Arthur  Kornberg. 
also  of  Stanford,  thinks  the  only  way  to 
interpret  the  Nobel  committee's  "care 
fully  worded  citation"  is  as  recognition 
for  Berg's  20  years  of  leadership  in  the 
molecular  biology  of  nucleic  acids. 

In  the  1960's.  Berg  did  a  great  deal  of 
ir:.ovative  work  on  bacterial  protein 
vnthesis.  particularly  the  interaction  of 
amino  acids  with  transfer  RNA's.  His 
work  helped  explain  how  these  RNA's 
are  used  as  adapters  in  decoding.  His 
group  and  several  others  also  discovered 
one  of  the  enzymes  that  copies  DNA  into 
RNA. 

Then,  about  10  years  ago.  Berg  and 

SCIENCE.  VOL.  :io.  :i  NOVEMBER  ivso 


many  other  molecular  biologists  became 
interested  in  applying  what  is  known 
about  bacterial  gene  expression  to  the 
study  of  gene  expression  in  higher  orga 
nisms.  "We  began  to  think  of  using  SV40 
[an  animal  tumor  virus]  to  carry  genes  in 
to  m.imma!i;:;i  colls,"  Bcr-  v.;<,.  Tin- 
foreign  genes  could  then  be  studied  and 
manipulated  to  see  what  controls  their 
expression. 

In  1971,  Berg  and  his  colleagues  David 
Jackson  and  Robert  Symons  opened  the 
circular  SV40  molecule  with  a  restriction 
enzyme,  Eco  Rl.  This  enzyme,  which 
w;as  discovered  in  Herbert  Boyer's  labo 
ratory  at  the  University  of  California  at 
San  Francisco,  cleaves  DNA  at  specific 
base  sequences.  In  the  case  of  SV40 
DNA,  it  cleaves  it  in  exactly  one  spot. 
Berg's  group  then  spliced  the  linear 
SV40  DNA  to  the  DNA  of  the  bacterial 
virus  \.  The  X  DNA  also  is  circular  and 
Berg's  group  cleaved  it  too  with  Eco  Rl. 

Although  this  was  the  first  time  that 
DNA's  from  two  different  species  were 
joined,  it  was  not  the  first  time  that  any 
DNA's  were  joined.  H.  Gobind  Kho- 
rana.  of  the  Massachusetts  Institute  of 
Technology,  discovered  in  the  1960's 
that  an  enzyme  produced  by  the  bacte 
rial  virus  T4  can  catalyze  the  linking  to 
gether  of  DNA  molecules.  Berg.  Jack 
son,  and  Symons  enzymatically  con 
structed  complementary  or  "sticky" 
ends  on  the  two  DNA  segments  to  be 
joined  and  then  used  the  T4  enzyme  to 
do  the  joining.  The  method  they  used 
was  developed  and  tested  independently 
by  Berg's  group  and  by  Peter  Lobban 
and  Dale  Kaiser  of  Stanford.  Although 
no  one  knew  it  at  the  time,  it  was  unnec 
essary  to  construct  sticky  ends,  since 
they  are  automatically  produced  when 
Eco  Rl  cleaves  DNA.  This  fact  was  dis-  Paul  Berg 

0035-8075,80' I  i:i-088?S00.50,'0     Copyright  C  1980  AAAS 


covered  in  1972  by  Janet  Mertz  and  Ron 
ald  Davis  and  independently  by  Vittorio 
Sgaramella.  all  of  Stanford  University. 

It  had  been  Berg's  intention  to  in 
troduce  the  SV40-A.  hybrid  molecule  into 
the  bacterium  Eschericliiu  cnli .  which  X 
c;m  infect.  In  th.'.t  wny.  he  could  pet  mnn\ 
copies  of  the  molecule  to  be  used  tor 
future  experiments  in  gene  expression  in 


88- 


woe  World  Pholo 


Frederick  Sanger 


mammalian  cells.  In  the  summer  of 
1971,  Mertz,  who  was  Berg's  graduate 
student,  described  the  plan  at  a  tumor  vi 
rus  conference  held  at  Cold  Spring  Har 
bor.  New  York.  Robert  Pollack  of  Cold 
Spring  Harbor  Laboratories  reacted  im 
mediately  with  dismay,  pointing  out  that 
SV40  transforms  human  cells  in  culture 
and  that  E.  coli  lives  in  the  human  gut.  If 
any  E.  coli  infected  with  the  SV40-X 
DNA  escaped  from  the  laboratory,  they 
could  be  dangerous. 

Berg  was  persuaded  by  this  argument 
and  decided  not  to  do  the  experiment. 
He  led  molecular  biologists  in  calling  for 
a  moratorium  on  recombinant  DNA  re 
search  until  the  risks  could  be  assessed 
.ind  the  safety  of  the  experiments  en 
sured.  It  was  a  period.  Berg  recalls,  "of 
more  controversy  than  science."  In 
19?5.  the  moratorium  w-as  conditionally 
lifted  and  the  National  Institutes  of 
Health  developed  guidelines  for  the  con 
duct  of  recombinant  DNA  experiments. 
The  guidelines  have  since  been  softened 
as  the  experiments  turned  out  to  be  less 
risky  than  anticipated. 

Ironically,  the  experiment  that  Berg 
originally  wanted  to  do  would  not  have 
succeeded,  and  no  one  at  the  time  would 
have  known  uny.  The  SV40-A  hybrid 
would  not  have  replicated  in  bacteria  be 
cause  Berg  inserted  the  SV40  genes  at  a 
site  now-  known  to  be  essential  for  \'s 
replication  and  thereby  interrupted  this 
site. 

In  fact,  the  heart  of  recombinant  DNA_ 
technology  is  not  just  gene  splicing  but 
also  gene  cloning.  It  is  necessary  to  find 
ways  to  get  foreign  genes  into  cells,  en 
sure  that  the  genes  are  expressed,  and 
then  select  for  the  cells  that  are  ex 
pressing  those  genes.  Cloning  techniques 
were  pioneered  by  Stanley  Cohen  and 
Annie  Chang  of  Stanford  University  and 


242 

Herbert  Boyer  and  Robert  Helling  of  the 
University  of  California  at  San  Fran 
cisco,  who,  in  the  early  )970's,  devel 
oped  a  plasmid.  which  is  a  small  piece  of 
extra-chromosomal  DNA,  that  could 
carry  foreign  genes  into  bacterial  cells. 
The  plasmid  contained  genes  that  made 
the  bacteria  resistant  to  the  antibiotic  tet- 
racycline,  so  that  the  cells  which  took  up 
the  plasmid  and  expressed  its  genes 
could  easily  be  selected. 

In  the  past  decade,  recombinant  DNA 
techniques  have  become  increasingly  so 
phisticated.  Berg  has  played  a  major  role 
in  these  developments.  Most  recently, 
he  and  others,  particularly  Daniel  Na 
thans  of  Johns  Hopkins  University  Med 
ical  School,  who  won  a  Nobel  Prize  for 
his  work  in  restriction  enzymes,  exten 
sively  studied  the  structure,  organiza 
tion,  and  replication  of  SV40  genes.  Af 
ter  constnicting  deletion  mutants  of 
SV40  that  have  proved  extremely  useful 
in  studies  of  SV40  gene  functions.  Berg 
went  back  to  his  original  idea  of  using 
SV40  to  introduce  genes  into  mammalian 
cells.  He  spliced  to  SV40  an  E.  coli  gene 
that  allows  cells  tc  use  xanthine  as  a  sub 
strate  in  nucleotide  synthesis.  Then,  in 
separate  experiments,  he  spliced  animal 
genes  for  globin.  histone.  or  the  enzyme 
dihydrofolate  reductase  to  this  hybrid 
SV'40  molecule.  When  the  SV40  carrying 
the  added  bacterial  and  animal  genes  was 
introduced  into  cultured  cells.  Berg 
could  pick  out  the  cells  that  were  trans 
formed  by  SV40  by  selecting  for  cells 
that  grow-  on  xanthine.  In  this  way.  Berg 
was  able  to  show  that  the  added  animal 
gciu-3  ai'c  c.xpiesscJ  in  cultured  cells. 

Dean  Hamer  and  Philip  Leder  of  the 
National  Institute  of  Child  Health  and 
Human  Development  have  also  used 
SV40  as  a  cloning  vector  in  cultured 
cells.  But.  says  Nathans.  "Clearly  the 
notion  that  you  could  construct  a  vector 
with  animal  viruses  was  Berg's  idea." 

An  important  aspect  of  .Berg's  work 
has  been  his  extraordinary  ability  to  de 
velop  methodologies.  For  example,  he 
was  the  first  to  use  nitrocellulose  binding 
assays  to  study  interactions  between 
proteins  and  nucleic  acids.  He  also  de 
veloped  the  nick  translation  method, 
which  is  used  to  make  isotopically  la 
beled  DNA  probes  and  is  central  to  cur 
rent  studies  of  gene  functions.  "His  style 
of  biochemistry  helped  set  the  standards 
in  the  nucleic  acid  field."  says  Nathans. 

The  second  half  of  the  chemistry  prize 
was  also  given  to  developers  of  method 
ologies.  Sanger  and  Gilbert  were  hon 
ored  for  their  discoveries  of  ways  to  se 
quence  DNA.  In  the  past  few  years, 
these  techniques  have  become  widely 
used  to  determine  amino  acid  sequences 


of  proteins  because  with  these  method 
it  is/easier  and  more  accurate  to  se 
quence  the  DNA  coding  for  proteins 
than  to  sequence  the  proteins  directly. 
The  techniques  are  also  used  to  deter 
mine  the  intervening  sequences  that  oc 
cur  in  eukaryotic  genes  and  the  se 
quences  that  occur  in  control  regions  of 
bacterial  DNA.  By  using  these  methodv 
molecular  biologists  hope  to  learn  whi.'i 
sequences  control  gene  expression  .1 
higher  organisms  and  how  they  do  so 
"DNA  sequences  are  the  basic,  under 
lying  structures  [of  molecular  biology]. 
There  is  nothing  more  primitive.  Your 
questions  are  ultimately  posed  there," 
says  Gilbert. 

Sanger  and  Gilbert  are  about  as  dif 
ferent  as  two  scientists  can  be.  and  they 
came  upon  their  sequencing  methods  by 
entirely  different  paths.  Sanger  is  qu  :. 
modest,  self-effacing;  Gilbert  is  much 
more  flamboyant.  Ted  Friedman  of  the 
University  of  California  at  San  Diego, 
who  spent  a  sabbatical  year  with  Sanger. 
says.  "If  you  talk  to  Sanger  and  do  not 
know  who  he  is.  you  would  think  he  is 
the  lab  caretaker.  If  you  allow  him  to.  he 
will  melt  into  the  woodwork."  George 
Brownlee  of  Oxford  University,  who  un 
til  recently  was  at  Cambridge  with  S..:  :- 
er.  adds,  "Sanger  certainly  doesn't  give 
himself  airs.  But  in  my  view,  he  ranks 
among  the  great  scientists  of  our  time." 

According  to  Friedman.  Sanger's  out 
standing  feature  is  his  "uncanny  belief 
and  knowledge  that  sequencing  can  be 
determined  by  very  simple  methods."  In 
the  1950's,  Sanger  studied  protein  se 
quencing  -it  .1  tirr;  when  no  one  l--y.v 
whether  all  proteins  of  a  particular  :;  pe 


Walter  Gilbert 


'have  the  same  sequences.  His  first  Nobel 
i  Prize  was  awarded  for  this  work.  Then 
\  ),e  attacked  the  problem   of  RNA  se- 
I  quencing.  developing   the   widely  used 
.'  fingerprinting  method.   About   10  years 
\  ago.  he  set  out  to  sequence  DNA.  even 
*-.  thoir-h  this  problem,  too.  was  consid 
ered  intractable. 

S.inger's  method  evolved  gradually 
from  more  than  one  line  of  attack  on  the 
problem.  In  the  early  1970's.  he  discov 
ered  the  plus-minus  sequencing  method, 
a  direct  precursor  of  the  method  he  uses 
today.  In  the  plus-minus  method  the  ob 
ject  is  to  obtain  a  set  of  nested  segments 
of  the  DNA  to  be  sequenced.  The  first 
segment  consists  of  the  first  nucleotide, 
.  the  second  consists  of  the  first  two  nucle 
otides.  the  third  of  the  first  three  nucle- 
otides.  and  so  on.  These  segments  are 
constructed  in  such  a  way  that  the  identi 
ty  of  the  last  nucleotide  of  each  nested 
segment  is  known.  Once  obtained,  the 
nested  segments  can  be  separated  ac 
cording  to  size  by  electrophoresis  on  an 
ultrathin  polyacrvlamide  gel.  The  sepa 
rated  fragments  can  be  delected  because 
e:'.:h  is  isotopically  labeled. 

ir.c  ke>  l\j  llic  pli.i-nuni;^  method  U 
obtaining  the  nested  segments.  Sanger 
constructs  them  by  synthesizing  them. 
He  separates  the  two  strands  of  the  DNA 
to  be  sequenced  and  then  makes  partial 
copies  of  one  of  those  strands.  To  ensure 
that  the  partial  copies  include  all  nested 
segments  and  that  the  terminal  nucle 
otide  of  each  segment  is  know-n.  Sanger 
m  ikes  the  copies  under  conditions  in 
which  one  nucleotide  is  limited  in  quanti 
ty  For  example,  he  provides  limited 
quantities  of  adenine  so  that  the  DNA 
copying  will  eventually  stop  because  of 
lack  of  adenine.  Then  all  of  the  copies 
made  will  end  just  before  an  adenine. 
and  the  next  nucleotide  of  each  of  the  re 
sulting  segments  is  adenine.  In  a  similar 
way.  Sanger  synthesizes  segments  end- 
is'.;:  before  each  of  the  other  three  DNA 
nucleotides. 

Sanger  has  since  improved  the  plus- 
minus  method  to  make  it  more  efficient. 
Instead  of  supplying  limited  quantities  of 
each  nucleotide.  he  supplies  derivatives 
of  the  nucleotides  that  cause  DNA  syn 
thesis  to  stop. 

In  contrast  to  Sanger.  Gilbert  did  not 
deliberately  set  out  to  sequence  DNA.  A 
highly  visible,  active  scientist  who  runs  a 
i;irge  laboratory.  Gilbert  has  worked  on  a 
"•ide  variety  of  problems  in  the  past  20 
years,  ranging  from  how  bacterial  genes 
are  organized  and  expressed  to  gene  con 
trol  in  higher  organisms  and  genetic  engi 
neering.  He  is  also  chairman  of  the  board 
and  cochairman  of  the  board  of  directors 
of  the  gene  splicing  firm  Biogen. 

:i  NOVEMBER   1980 


Gilbert,  working  with  Allajn  Maxam. 
who  is  now  at  Harvard  MedicaJ  School's 
Sidney  Farber  Cancer  Institute,  came 
upon  a  DNA  sequencing  technique  aJ- 
most  by  chance.  Gilbert  recalls  that  one 
day  in  early  1975.  Andrei  Mirzabekov,  of 
the  USSR  Academy  of  Sciences,  ap 
peared  in  his  office  and  urged  him  to  try  a 
new  approach  to  studying  how  proteins 
recognize  specific  sequences  of  DNA. 
Gilbert  had  long  been  interested  in  the 
lac  represser  protein  of  £.  coli.  which 
binds  to  the  lac  operon  segment  of  DNA. 
and.  in  fact,  it  was  Gilbert  who  isolated 
the  lac  represser  and  operator.  Mirzabe 
kov  and  his  colleagues  had  been  probing 
protein-DNA  interactions  with  dimethyl 
sulfate.  a  reagent  that  methylates  the 
DNA  nucleotides  adenine  and  guanine. 
After  reacting  with  dimethyl  sulfate. 
DNA  breaks  easily  at  these  bases. 

Gilbert  decided  to  expose  lac  operon 
DNA  to  dimethvl  sulfate  and  then  break 


.  c 


« 1 


6- 

• 

T 


«• 
T 


^    •  • 


<     -• 

C 

c 


C.    **** 


Purl  of the  sequencing  pattern  obtained  from 
a  piece  of  Di\A  about  130  base  pairs  in 
length.  The  letters  at  the  top  of  the  columns, 
A.  C,  C,  and  T  (adenine,  guanine,  cyto- 
sine,  and  thymine)  indicate  which  base'*as 
preferentially  cleaved  by  chemicals.  Tue  dark 
est  band  in  each  column  represents  the  base 
missing  from  the  end  of  the  initial  segments, 
with  the  exception  of  cytosine  (C).  All  dark 
bands  in  the  C  column  represent  cytosines 
e\en  if  bands  also  appear  in  the  T  column  at 
that  position.  Bands  that  appear  in  the  T 
column  but  not  in  the  C  column  represent  thy- 
mines  IT>.  To  read  the  sequence  of  the  DNA  . 
read  off  the  base  represented  by  each  bund, 
sinning  from  the  bottom  of  the  columns. 
[Source:  Wiiller  Gilbert  and  Allan  Mu.ram] 


the  DNA  at  adenines  and  guanines.  For 
comparison,  he  would  bind  lac  represser 
to  the  operon  and  repeat  the  experiment. 
The  adenines  and  guanines  that  reacted 
with  the  represser  should  be  protected 
from  the  dimethyl  sulfate,  and  so  the 
DNA  should  not  break  there.  Since  the 
sequence  of  lac  operon  DNA  was  known 
(it  had  been  copied  into  RNA  and  the 
RNA  sequenced).  it  would  be  possible  to 
learn  where  the  represser  binds  on  this 
DNA. 

After  these  experiments.  Gilbert  and 
several  of  his  associates  discovered  a 
second  lac  operon  of  unknown  se 
quence.  Maxam  repeated  the  dimethyl 
sulfate  experiments  with  this  new  lac  op 
eron  and  the  lac  represser.  When  he  and 
Gilbert  saw  the  results,  they  realized  that 
they  had  the  beginning  of  a  DNA  se 
quencing  method.  By  using  dimethyl  sul 
fate  and  adjusting  the  reaction  condi 
tions,  they  could  break  DNA  at  either 
adenines  or  guanines.  Now  if  they  could 
find  a  way  to  break  DNA  preferentially 
at  thymines  or  cytosines,  they  could  gen 
erate  nested  segments  whose  terminal 
nucleotides  were  known.  With  this  idea. 
Maxam  set  to  work  to  develop  ways  of 
breaking  DNA  at  tli>mini.-b  c:  c>iu>ine>. 
He  recalled  that  under  appropriate  chem 
ical  conditions,  hydrazine  preferentially 
weakens  DNA  at  one  or  the  other  of 
these  nucleotides.  After  a  summer  of 
work.  Maxam  succeeded  in  perfecting 
the  chemical  method  of  sequencing 
DNA. 

The  difference  between  the  Sanger 
method  and  the  Maxam-Gilbert  method 
is  that  Sanger  generates  nested  segments 
by  synthesizing  them  and  Maxam  and 
Gilbert  generate  the  segments  by  break 
ing  the  DNA  at  specific  bases.  Both 
methods  are  currently  used,  and  re 
searchers  experienced  with  both  say  that 
the  choice  between  them  depends  in  part 
on  the  length  of  DNA  to  to  be  sequenced 
and  in  part  on  the  personal  preferences 
of  the  investigator.  Tom  Maniatis  of  the 
California  Institute  of  Technology,  for 
example,  uses  Sanger's  method  for  very 
long  sequences  of  DNA  because  it  is 
faster.  For  shorter  sequences,  one  or  a 
fey  genes  long,  the  two  methods  are 
comparable  in  speed,  but  Maniatis  pre 
fers  the  Maxam-Gilbert  method  because 
"Allan  has  established  the  protocol  so 
completely  that  anyone  who  tries  the 
method  is  successful." 

The  full  ramifications  of  recombinant 
DNA  technology  and  DNA  sequencing 
methods  are  not  yet  known.  But  these 
techniques  are  changing  molecular  biolo 
gists'  perceptions  of  what  can  be  learned 
about  the  genes  of  higher  organisms. 

_GINA  BARI  KOLATA 

889 


INDEX--Paul  Berg 


244 


Abraham  Lincoln  High  School,   4-7 
Albert  Einstein  College  of 

Medicine,   172 
ALZA  [Corporation],   138,  149, 

152,  157,  161-162 
American  Cancer  Society,   32,  33 

Scholar  in  Cancer  Research,   39 
Amgen,   142 
amino  acid  assembly/activation, 

43-45.   See  also  protein 

synthesis /assembly 
antibiotic  resistance,   77 
anti-Semitism,   8,  15 
Arai,  Kenichi,   152,  163-165 
Arrowsmith,   3 

artificial  kidney  research,   17 
Asilomar,   136,  185 
Asilomar  1  [Conference  on 

Biohazards  in  Biological 

Research],   70,  72-73,  75-76, 

92-94,  115,  120,  161,  185 
Asilomar  II  [Conference  on 

Recombinant  DNA] ,   72,  74,  76, 

77-79,  121 


bacteriophage,   32,  54,  60-61,  88, 

98 

as  transducing  agents,   64-65 

complimentary  tails,   66-67,  89 

lambda,   52-53,  64-66,  68-69, 
82,  88,  90,  103,  108 

pi,   84-85 

p22,   108,  111 

phiSO,   65,  82,  88 

T-4,   60 

X,   59 

Baldwin,  Robert  "Buzz",   175 
Baltimore,  David,   71,  74,  120, 

138,  147 

Beadle,  George,   61 
Beckman  Center  for  Molecular  and 

Genetic  Medicine.   See  Stanford 

University 


Beckman  Foundation,   178 
Beckman,  Arnold,   181-183 
Benzer,  Seymour,   51 
Berg,  Irving,   5 
Berg,  Jack,   5 
Berg,  Millie,   13 
Biochemical  and  Biophysical 

Research  Communications,   129- 

130 
Biochemistry  Department,  Stanford, 

90.   See  also  Stanford 

University 
Biogen,   159-160 
biohazards/biosafety ,   71-80 

NIH,   74 

Salk  Institute,   72-73 

Watson  stance,   76 

See  also  Asilomar  I  and  II 
Bishop,  Mike,   57,  176,  179 
Bodmer,  Walter,   173-174 
Bohr,  Niels,   28-29 
Bollum,  Fred,   89,  128 
Boyer,  Herbert,   70-71,  77-78,  95, 

99,  101-102,  105,  112-113,  117- 

118,  120-122,  126,  139-141 
Brenner,  Sydney,   156 
Brodsky,  Sarah  (mother),   1 
Broker,  Tom,   125 
Bronx  High  School  of  Science,   6 
Brooklyn  College,   9-10 
Brooklyn  Tech,   9 
Brown,  Don,   87,  114 
Brown,  Mike,   179 


Calgene,  144 
Caltech,  177 
Cambridge  Medical  Research 

Council,   57,  63 
Cantor,  Harvey,   163 
Cape,  Ronald,   141 
Carnegie  Labs,  Johns  Hopkins,   87, 

114 
Cavalli-Sforza,  Luca,   174 


245 


Cetus  [Corporation],   117,  141- 

142,  144-145 
Chamberlain,  Mike,   45 
Chemical  and  Engineering  News,   25 
Ciba-Geigy,   157 
City  College  of  New  York,   8 
cloning  (DNA) .   See  recombinant 

DNA  method 
Cohen,  Stanley,   110,  126,  172-174 

recombinant  DNA  method,   69-71, 
77-78,  101,  105-106,  112-122 

relation  to  Stanford 

biochemistry  dept.,   90,  95, 
116-119,  142,  144,  181,  183 
Cohn,  Melvin,   51-52 
Cold  Spring  Harbor  Laboratory, 

76,  88,  91,  98,  125 
Congressional  training  grants, 

134-135 
consulting  in  academia,   138,  143- 

148 
Cori,  Carl,   25-27,  30,  34,  40,  58 

Nobel  Prize  with  wife,  Gerty, 

27,  124 

Cornell  Medical  School,   19 
Crabtree,  Jerry,   183 
Crick,  Francis,   33,  44,  46,  48, 

57 

Cuzin,  Francois,   59,  99 
cytokines,   163-164,  166-168 


d'Andrade,  Hugh,   160 

Danish  Royal  Academy  of  Sciences, 

28-29 
Danish  State  Serum  Institute 

(Copenhagen) ,   32 
Davis,  Ronald,   71,  100-102,  120- 

122 

de  Kruif,  Paul,   3 
Dieckmann,  Marianne,   59 
Djerassi,  Carl,   132-133,  142 
DNA 

replication,   46-51,  54,  60-61, 
66,  71,  89,  90,  112 

structure,   33 

synthesis,   46-48,  68,  95,  96 

transcription/translation,   45, 
51,  60,  84-85 


DNAX  Research  Institute,   138-139, 
149-169.   See  also  Schering 
Plough 

Drosophila  genetics,   59,  61,  63, 
70,  112 

du  Pont,   133 

du  Vigneaud,  Vincent,   19,  22 

Dulbecco,  Renato,   59 

Dynapol,   162 


enzyme /enzymology,   21,  62-63 
activation,   51-52 
enzyme  reactions,   26,  29,  30, 

36-37,  42,  89 
Kalckar,   41 

Kornberg,   21,  25,  40-41 
ligase,   67-68,  96-97,  108, 

130,  175 

nucleic  acid,   21,  35 
purification,   25,  36-37,  41- 

42,  44-45,  47-48,  57,  61, 

63,  87 
terminal  transf erase,   111, 

128-129 


fatty  acid  activation,   38,  42 
Federation  for  Experimental 

Biology  meetings,   38 
Fredrickson,  Don,   180-182 
Friedman,  Ted,   92 


G.I.  Bill,   19 
Ganeson,   174 
Gellert,  Martin,   67 
gene 

expression,   51-52,  54-55,  58, 
60,  66,  71,  84-85,  104,  155, 
164 

regulation,   51-53,  55,  58,  85, 
155 

repression,   52-54 

selection,   83 

therapy,   74-75,  104 

transfer,   82,  84,  86 
Genentech  Inc.,   117,  139-141 
General  Foods  Corporation,   13 


246 


genetic  engineering,   75,  81,  123, 

136,  138,  154,  162 
Genetics  Institute,   164,  166 
germ  warfare,   91,  94 
Gilbert,  Wallace,   126 
Glaser,  Don,   141-143 
Goldstein,  Joe,   179 
Gordon  conference,   119-120 
Goulian,  Mehran,   131 
graduate  student  teaching,   18-19 


Haber,  Edward,   152-156 
Hall,  Eliza  Institute,   173 
Harvard,   43,  45,  99,  107,  151- 

152,  163 

Hayaishi,  Osamu,   34 
Helling,  Robert,   115-116 
Hershey,  Alfred,   67,  88 
Hertzenberg,  Leonard,   173 
Hoagland,  Mahlon,   43-45 
Hogness,  David,   51-52,  59,  70-71, 

112,  120-122 
Hood,  Lee  153,   164 
hoof  and  mouth  disease,   77 
Hughes,  Howard,   183 

Medical  Institute  at  Stanford, 

180-184 
Hurwitz,  Jerry,   112-113,  172 


ICRF,   57 

Immunex,   166 

immunology,   152,  157,  160-163, 

165-168,  174,  178 
Industrial  Affiliates  Program 

[IAP],  Stanford,   132-137 
Institute  for  Research  on  Aging, 

Stanford,   134 


Jackson,  David,   69,  72,  92-95, 
97,  102-103,  107,  109,  111,  131 

Jacob,  Francois,   52,  55,  58-59 

Jensen,  R.H.,   110-111,  123,  129- 
131 

Joklik,  William,   29 


Kaiser,  Dale,   51-53,  56,  59,  66, 

68,  88,  103,  107,  114,  133, 

137,  174 

Kalckar,  Herman,   24-33,  41 
Kaplan,  Henry,   146 
Kennedy  Foundation,   175 
Kennedy,  Donald,   144,  182 
Kenyon,  Cynthia,   63 
Kern,  Dave,   182 
Khorana,  Gobind,   57,  96-98,  127, 

175 

Kolff ,  Willem  J. ,   17 
Koprowski,  Hilary,   110 
Kornberg,  Arthur,   5 

academic  background,   40-41 

ALZA/DNAX,   149-169 

apprenticeship  with  Cori,   27 

City  College,   7 

consulting  fees,   146 

DNA  polymerase  I,   89,  96,  128, 
131 

DNA  replication,   46-51 

DNAX,   139 

E.  coli  in  research,   56-58 

enzyme  access  at  Stanford,   67 

enzymology,   21,  25 

high  school,   6-7,  25 

meeting  Berg,   24 

nearest  neighbor  experiment, 
48 

NIH,   27-28,  34,  40-41 

Nobel  Prize,   6,  123,  125-127 
Committee,   125,  126 

Paul-Lewis  Award,   25,  41 

professor  of  microbiology  at 
Washington  Univ.,  27-28, 
33-36,  38-39,  46 

Senate  testimony,   75 

Stanford  biochemistry  IAP,   133 

Stanford  departments,   174, 

182,  185 

Kornberg,  Roger,   57 
Kornberg,  Tom,   63 
Kornberg,  Sylvy,   34 


Lear,  John,   102 

Lederberg,  Joshua,   75,  81,  91-92, 
141-142,  171-174 


247 


Lehman,  Bob,   67,  89,  137 
Lennette,  Edwin,   78 
Leonard,  Jack,   17 
Levy,  Mildred,   4 
Lewis,  Andy,   75,  78 
Lewis,  Sinclair,   3 
Lhu,  Gilbert,   176 
Lieberman,  Irving,   34 
Lipmann,  Fritz,   35-36,  38 
Lipton  Tea  Company,   13 
Lobban,  Peter,   68-69,  102-111, 

122,  128,  131 
Lowry,  Oliver,   34 
Luciano,  Robert  P.,   159,  167 
Luria,  Salvador,   32,  33 
Lwoff,  Andre,   52-53,  58 
Lyman,  Richard  W. ,   143 
Lynen,  Feodor,   35-36,  38 
lysogeny,   53-54,  80-86,  88 


Maaloe,  Ole,   32 

Mack,  [Senator]  Connie,   57 

mammalian  cell  biology,   60,  65- 

66,  69,  74,  77,  83-87,  90,  104- 

105,  107,  118,  119 
Mandel,  Mort ,   114,  173-174 
Maniatis,  Tom,   154 
Markey  Fellowship,   39 
Massachusetts  Institute  of 

Technology  [MIT],   70,  98,  147, 

149,  177 

biohazards  meeting,   74,  76, 

120-121,  161 
Matsubara,  Kenichi,   66 
McDevitt,  Hugh,   142,  144,  181 
Melman,  Kenneth,   179,  181,  183 
Merck,   111-112 
Mertz,  Janet,   91,  98-102,  114, 

116,  173-174 
Meselson,  Matthew,   99 
messenger  RNA,   44-45,  49-51,  60, 

84-85 

Meyers,  Victor,   16,  17 
Microbe  Hunters,   3 
monoclonal  antibodies,   153-156 
Monod,  Jacques,   52,  55,  58 
Moore,  Kevin,   153,  164 


Morrow,  John,   99-102,  105-106, 

112,  114-116,  119-120,  128 
Mullis,  Gary  127 
mutations,   60-64,  74,  104,  127 


Nathans,  Daniel,   71 
National  Academy  of  Sciences, 

119-121 
National  Institutes  of  Health 

[NIH],   27,  28,  32,  40,  57,  75, 

180 
New  York  City 

Brighton  Beach,   2 

Brooklyn,   2,  25 

Coney  Island,   2 
Nobel  Prize 

Berg,   6,  69 

acceptance  speech,   117, 
122-127 

Jerome  Karle,   6 

Gobind  Khorana,   57 

Arthur  Kornberg,   6 

Fritz  Lipmann,   35 

Vincent  du  Vigneaud,   19 
Nobel,  Alfred,   127 
Nossal,  Gustav  J.V.,   174 
nucleic  acid  biology/biochemistry, 

29-30,  35-38,  46,  50,  57,  85, 

89,  96 
nutrition  research,   12,  19-21,  42 

nutritional  supplement 
research,   19-20 


Oak  Ridge  National  Laboratory, 

22,  23 
Ochoa,  Severe,   40,  57,  123,  185- 

186 

Ofengand,  James,   43 
Okayama,   154-155,  164-165 
Oklahoma  A  and  M  (Oklahoma  State 

University),   15-16 
Omura,   156 
oncogenesis,   54,  57,  73,  77 


Pasteur  Institute,   51-52,  59 
patenting,   115-116 


248 


Paul-Lewis  Award  in  Enzyme 

Chemistry  (Berg),   41 
Pennsylvania  State  University, 

10-14 

plasmidology,   95,  112,  115 
Plum  Island,   77 
polio  vaccine,   56,  93 
Pollack,  Bob,   91-92 
polymerase  chain  reaction  [PCR] , 

127 
postdoctoral  positions  (Berg) 

Copenhagen,   26 

St.  Louis  (Washington 

University),   26-28 
protein  synthesis,  assembly,   42- 

45,  49-51,  51-52,  64 
Public  Health  Research  Institute 

in  New  York,   25 
Purpura,  Dominick,   181 


radioisotopes ,   14-15,  18,  22-24, 

46-47 
recombinant  DNA 

biohazards  controversy,   56, 
71-81,  91-94,  119-122,  125- 
126,  139,  179 
"Berg"  moratorium  letter, 

71,  74,  76,  120-122,  161 
James  Watson,   76,  120-121 
commercialization  of 

biomolecular  science,   116- 
117,  132-153 

commercial  reagents,   60 
See  also  DNAX 

method,   69-71,  77-78, 
101,  105-106,  112-122 
regulatory  policy,   81 
restriction  enzymes,   70,  94- 
95,  99,  113 
EcoRl,   99-102,  112-114, 

116,  119,  128-129,  173 
science,   64-90,  92-118,  121- 
131,  136-137,  140-142,  152, 
162 

cohesive  ends,   66-67,  88- 
90,  94-95,  98-103,  108, 
111,  113,  129,  173 
See  also  Asilomar  II 


ribosoraes,   44,  87 
Roberts,  Richard,   125 
Robertson,  Channing,   150-152 
Roblin,  Richard,   71,  74 
Rogoff,  M.H.,   110 
Roosevelt,  President  Franklin 

Delano,   7 

Rosenberg,  Thomas,   29-30 
ROTC  [Reserve  Officer  Training 

Corps],   9 

Rothchild  [Venture  Capital],   156 
Rutter,  William  J.,   140-141 


Sakami,  Warwick,   20 

Salk  Institute,   59,  65,  84-85 

Salk,  Jonas,   56 

Sanger,  Eugene,   126 

Schering-Plough,   157-163,  167- 

168.   See  also  DNAX 
Schodnick,  Gary,   183 
Science  for  the  People,   80 
Sgaramella,  Vittorio,   96-98,  102, 

108,  122,  172,  175 
Sharp,  Phil,   125 
Shooter,  Eric,   175-176 

Shooter  Committee  on  Conflicts 
of  Interest,  Stanford,   145- 
148 

Silicon  Valley,   134 
Singer,  Maxine,   61,  120,  161 
Skaggs,  Leonard,   17 
smallpox,  laboratory  outbreak,   80 
Smith,  Michael,   127 
SmithKline  Beecham,   182 
space  biology,   173-174 
Spudich,  Jim,   177 
Stanford  University,   52,  58-59, 

66-67,  72-73,  88-89,  94,  96-98, 

112,  129,  132-137,  140,  142, 

144,  167-187 

Beckman  Center  for  Molecular 
and  Genetic  Medicine,   62, 
110,  177-185 

Berg  achievements,   185-187 

biochemistry  dept.,   172-176 

genetics  dept.,   171-175 

Howard  Hughes  Medical 
Institute,   180-184 


249 


Stanford  University  (cont'd.) 

Industrial  Affiliates  Programs, 
132-137 

Program  in  Molecular  and 

Genetic  Medicine,   184 
Stark,  George,   57 
SV40  [Simian  Virus  40],   54-56, 

59,  64-65,  69,  71-72,  75-77, 

85-87,  89-90,  92-93,  95,  97- 

100,  104,  107-109,  119 

genome,   60,  65-66,  68,  76,  117 
Swanson,  Robert,   140 
Swedish  Royal  Academy,   122,  127 
Symons,  Robert  H. ,   69,  93,  102, 

109,  129,  131 
Syntex,   142,  158 


Takeda  Chemical,   156 
Tokyo  University,   165 
transduction 

transduction  system,   65-66, 

80-86,  102,  104,  119 
agents,   64-65 
transfer  RNA  (tRNA),   43-45,  49- 

51,  64 
tumor  virus  research 

mammalian,   53-61,  65,  72,  75- 

77,  84-85,  91-94,  120,  164 
polyoma,   54-56,  65,  84-85 
SV40  [Simian  Virus  40],   54-56, 
59,  64-65,  69,  71-72,  75-77, 
85-87,  89-90,  92-93,  95,  97- 
100,  104,  107-109,  119 
genome,   60,  65-66,  68,  76, 

117 

Ultimate  Experiment,   71 
University  of  California,   144 
UC  Berkeley,   45 
UC  Davis,   144 
UCSF,   63,  99,  117,  139-141, 
176-178 
Program  in  Biological 

Sciences,   176,  178 
University  of  Michigan,   72,  92 


Vietnam  War,   80 


Wade,  Nicholas,   71,  94 

Waitz,  J.  Allan,   159-160 

Wang,  Jim,   107 

Washington  University  (St.  Louis), 

26-28,  33-34,  39,  51-52,  170 
Watson,  James  D. ,   32-33,  44,  46, 

48,  71,  74 

biohazard  position,   76,  120- 

121 

Weiner,  Charlie,   149 
Weissman,  Irving,   183 
Weissman,  Sherman,   71 
Western  Reserve  Medical  Center, 

19 
Western  Reserve  University,   14-24 

Department  of  Biochemistry, 
17-26 

Department  of  Clinical 

Biochemistry,   16-17,  20 
Wodzinski,  R.J.,   110 
Wolf,  Sophie,   5-7 
Wood,  Bill,   45 

Wood,  Harland,   15-16,  18,  20-27 
World  War  II,   7 
-  Hiroshima,   12 

occupation  and  rationing  in 
Denmark,   31 

Pearl  Harbor,   7 

radiation  labs,   22-23 

service,   11-12 
Wright,  Barbara,   32 
Wright,  Susan,   81 


Yanofsky,  Charles,   64,  69,  82, 
102,  107,  139,  149-153 


Zaffaroni,  Alejandro,   138,  142, 

150-158 

Zamecnik,  Paul,   43-45 
Zinder,  Norton,   71,  74,  81 
Zurowski,  Gerard,   153,  64-165 


Valentine,  Ray,   144 
Varraus,  Harold,   179 


Sally  Smith  Hughes 


Graduated  from  the  University  of  California,  Berkeley,  in 
1963  with  an  A.B.  degree  in  zoology,  and  from  the  University 
of  California,  San  Francisco,  in  1966  with  an  M.A.  degree  in 
anatomy.   She  received  a  Ph.D.  degree  in  the  history  of 
science  and  medicine  from  the  Royal  Postgraduate  Medical 
School,  University  of  London,  in  1972. 

Postgraduate  Research  Histologist,  the  Cardiovascular 
Research  Institute,  University  of  California,  San  Francisco, 
1966-1969;  science  historian  for  the  History  of  Science  and 
Technology  Program,  The  Bancroft  Library,  1978-1980. 

Presently  Research  Historian  and  Principal  Editor  on  medical 
and  scientific  topics  for  the  Regional  Oral  History  Office, 
University  of  California,  Berkeley.   Author  of  The  Virus:  A 
History  of  the  Concept,  Sally  Smith  Hughes  is  currently 
interviewing  and  writing  in  the  fields  of  AIDS  and  molecular 
biology /biotechnology . 


2495