================================================================================

                       SECURITIES AND EXCHANGE COMMISSION
                             Washington, D.C. 20549
                              ---------------------

                                    FORM 10-K
                              ---------------------

                                   (Mark One)
                  ANNUAL REPORT PURSUANT TO SECTION 13 OR 15(D)
                     OF THE SECURITIES EXCHANGE ACT OF 1934
                   For the fiscal year ended December 31, 2000
                                       OR
                TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(D)
                     OF THE SECURITIES EXCHANGE ACT OF 1934
                        For the transition period from to

                         Commission file number: 0-28150

                          NEUROCRINE BIOSCIENCES, INC.
             (Exact name of registrant as specified in its charter)

- --------------------------------------------------------------------------------

                  Delaware                                 33-0525145
        (State or other jurisdiction                    (I.R.S. Employer
     of incorporation or organization)               Identification Number)
- --------------------------------------------------------------------------------
- --------------------------------------------------------------------------------

 10555 Science Center Drive, San Diego, CA                   92121
  (Address of principal executive office)                  (Zip Code)
- --------------------------------------------------------------------------------

       Registrant's telephone number, including area code: (858) 658-7600

        Securities registered pursuant to Section 12(b) of the Act: None
           Securities registered pursuant to Section 12(g) of the Act:
                         Common Stock, $0.001 par value

         Indicate  by check  mark  whether  the  registrant:  (1) has  filed all
reports  required to be filed by Section 13 or 15(d) of the Securities  Exchange
Act of 1934 during the preceding 12 months (or for such shorter  period that the
registrant was required to file such reports),  and (2) has been subject to such
filing requirements for the past 90 days.  Yes [X]  No [   ]

         Indicate by check mark if disclosure of delinquent  filers  pursuant to
Item 405 of Regulation S-K is not contained  herein,  and will not be contained,
to the best of the  registrant's  knowledge,  in definitive proxy or information
statements  incorporated  by  reference  in Part  III of this  Form  10-K or any
amendment to this Form 10-K. [X]

         The aggregate  market value of the voting stock held by  non-affiliates
of the  Registrant  as of March  23,  2001  totaled  approximately  $344,192,047
million  based on the closing  stock  price as  reported by the Nasdaq  National
Market.  As of March 23, 2001, there were 25,428,731  shares of the Registrant's
Common Stock, $.001 par value per share, outstanding.

                       DOCUMENTS INCORPORATED BY REFERENCE

         Certain  information  required by Part III of Form 10-K is incorporated
by reference  from the  Registrant's  Proxy  Statement for the Annual Meeting of
Stockholders to be held on May 24, 2001 (the "Proxy  Statement"),  which will be
filed with the  Securities  and  Exchange  Commission  within 120 days after the
close of the Registrant's fiscal year ended December 31, 2000.


================================================================================







                                TABLE OF CONTENTS

                                                                       Page
PART I
Item 1.     Business ................................................    3
Item 2.     Properties ..............................................   31
Item 3.     Legal Proceedings .......................................   31
Item 4.     Submission of Matters to a Vote of Security Holders .....   31

PART II
Item 5.     Market for the Registrant's Common Equity
               and Related Stockholder Matters ......................   31
Item 6.     Selected Financial Data .................................   32
Item 7.     Management's Discussion and Analysis of
               Financial Condition and Results of Operations ........   33
Item 7A.    Quantitative and Qualitative Disclosures on Market Risk .   38
Item 8.     Financial Statements and Supplementary Data .............   38
Item 9.     Changes in Disagreements with Accountants on
               Accounting and Financial Disclosure ..................   38

PART III
Item 10.    Directors and Executive Officers of the Registrant ......   39
Item 11.    Executive Compensation ..................................   39
Item 12.    Security Ownership of Certain Beneficial Owners
               and Management .......................................   39
Item 13.    Certain Relationships and Related Transactions ..........   39

PART IV
Item 14.    Exhibits, Financial Statements and Schedules
               and Reports on Form 8-K ..............................   39







                                     PART I

                           Forward-Looking Statements

         This Annual  Report on Form 10-K and the  information  incorporated  by
reference contain forward-looking  statements that involve a number of risks and
uncertainties.  Although our  forward-looking  statements reflect the good faith
judgment  of our  management,  these  statements  can only be based on facts and
factors  currently  known by us.  Consequently,  forward-looking  statements are
inherently subject to risks and  uncertainties,  and actual results and outcomes
may differ materially from results and outcomes discussed in the forward-looking
statements.

         Forward-looking   statements   can  be   identified   by  the   use  of
forward-looking  words such as "believes,"  "expects,"  "hopes,"  "may," "will,"
"plan," "intends," "estimates," "could," "should," "would," "continue," "seeks,"
"pro forma" or "anticipates," or other similar words (including their use in the
negative),  or by discussions  of future matters such as the  development of new
products,  technology  enhancements,  possible  changes in legislation and other
statements that are not historical. These statements include but are not limited
to statements  under the captions "Risk Factors,"  "Management's  Discussion and
Analysis of Financial  Condition and Results of  Operations"  and  "Business" as
well as other  sections in this report.  A number of factors could cause results
to differ materially from those anticipated by the  forward-looking  statements,
including those discussed under "Risk Factors" and "Management's  Discussion and
Analysis of Financial  Condition and Results of Operations." You should be aware
that the  occurrence  of any of the events  discussed  under "Risk  Factors" and
elsewhere in this prospectus could  substantially harm our business,  results of
operations and financial  condition and that if any of these events occurs,  the
trading price of our common stock could decline and you could lose all or a part
of the value of your shares of our common stock.

         The  cautionary  statements  made in this  report  are  intended  to be
applicable to all related forward-looking statements wherever they may appear in
this report.  We urge you not to place undue reliance on these forward-  looking
statements, which speak only as of the date of this report.


ITEM 1.  BUSINESS

Overview

            Neurocrine  Biosciences,  Inc. is a product-based  biopharmaceutical
company focused on neurologic and endocrine diseases and disorders.  Our product
candidates  address  some of the  largest  pharmaceutical  markets  in the world
including insomnia, anxiety, depression,  cancer and diabetes. We currently have
15 programs in various stages of research and development. Of these 15 programs,
five  programs are in clinical  development  and three  programs are in advanced
preclinical  development  which we expect to progress into human clinical trials
in the near  future.  We believe the other  seven  research  projects  will help
supply clinical  development  candidates in the future.  While we  independently
develop the majority of our product candidates, we utilize collaborators in four
of our 15 programs,  including Janssen Pharmaceutica,  a subsidiary of Johnson &
Johnson, Wyeth-Ayerst Laboratories, a division of American Home Products, Taisho
Pharmaceutical and Eli Lilly. Under these  collaborations,  we receive funds for
product  development,  in addition to receiving milestone payments and royalties
on worldwide product sales.


Our Business Strategy

            Our  goal  is  to  become  the  leading  therapeutic   product-based
biopharmaceutical  company  focused on  neurologic  and  endocrine  diseases and
disorders. There are six key elements to our business strategy:

            Build a Large and Diversified  Product Portfolio to Mitigate Overall
Clinical  and  Technical  Risk.  We believe that by building a large and diverse
product  pipeline,  we can  mitigate  some of the  risks  associated  with  drug
development.  We  currently  have 15 programs in various  stages of research and
development  with five  projects  in  clinical  development,  three  programs in
advanced preclinical  development which we expect to progress into the clinic in
the near future,  and seven research  projects to supply clinical  compounds for
the future. We take a portfolio  approach to managing our pipeline that balances
the size of the  market  opportunities  with  clear  and  defined  clinical  and



regulatory  paths to  approval.  We do this to ensure that we focus our internal
development  resources  on  innovative  therapies  with  high  probabilities  of
technical and commercial success.

          Identify  Novel  Drug  Targets  for  the   Development  of  Innovative
Therapies   to  Address   Large  Unmet  Market   Opportunities.   We  utilize  a
multidisciplinary  research approach to identify and validate novel drug targets
for internal development or collaboration. For example,  corticotropin-releasing
factor,  which is  believed  to be the  central  mediator  of the body's  stress
response, may represent the first new breakthrough for anxiety and depression in
over 20 years.  Gonadotropin-releasing factor antagonists, compounds designed to
reduce the  secretions  of sex  steroids,  may  represent the first novel orally
available means of treatment of prostate cancer and endometriosis.  Melanocortin
and  hypocretin  modulators are  compounds,  which affect  proteins in the brain
believed  to be  involved  in many  activities  of the body.  We  believe  these
compounds build upon our franchise and expertise in obesity and sleep disorders.
The creativity and productivity of our discovery research group will continue to
be a critical component for our continued success. Our team of approximately 150
biologists  and  chemists has a goal of  delivering  three  innovative  clinical
compounds  over  the  next  five  years to fuel  our  research  and  development
pipeline.

         Establish Corporate Collaborations with Global Pharmaceutical Companies
to Assist in the  Development of Our Products and Mitigate  Financial Risk While
Retaining Significant Commercial Upside. We leverage the development, regulatory
and commercialization expertise of our corporate collaborators to accelerate the
development of our potential  products while typically  retaining  commercial or
co-promotional  rights  in North  America.  We intend to  further  leverage  our
resources  by  continuing  to enter into  strategic  alliances  to  enhance  our
internal development and commercialization  capabilities.  Our current strategic
alliances include:

o        Janssen,   to  focus   on   corticotropin-releasing   factor   receptor
         antagonists to treat anxiety and depression;

o        Wyeth-Ayerst, to research, develop and commercialize compounds to treat
         neurodegenerative and psychiatric diseases;

o        Taisho,  to develop our  compound to treat Type 1 Diabetes in which the
         body does not produce enough insulin; and

o        Eli  Lilly,   to  collaborate  in  the   discovery,   development   and
         commercialization  of treatments of central  nervous system  disorders,
         including obesity.

         Acquire Rights to Complementary Drug Candidates. We plan to continue to
selectively  acquire rights to products in various stages of development to take
advantage of our drug  development  capabilities.  In May 1998, we licensed from
the  National  Institutes  of  Health  an  interleukin  4 fusion  toxin  that is
currently in clinical  trials for recurrent  malignant  glioma.  In May 1998, we
acquired  Northwest  NeuroLogic,  Inc.  and thereby  considerably  expanded  our
research  pipeline.  Through this  acquisition,  we acquired the  technology and
intellectual property rights surrounding  excitatory amino acid transporters,  a
portion of which is now in  collaboration  with  Wyeth-Ayerst.  We also acquired
from  Northwest  NeuroLogic   melanocortin  technology  and  other  intellectual
property that we are developing.  In June 1998, we licensed exclusive  worldwide
commercial  rights for  NBI-34060,  our compound for the  treatment of insomnia,
from DOV  Pharmaceuticals  and have since  moved  this  compound  into  advanced
clinical development.

         Supplement Our Internal  Research  Capabilities by  Collaborating  with
Leading  Platform  Technology  Companies.  We  believe  we  can  complement  our
multidisciplinary  research  process by selectively  accessing new  technologies
from  platform  technology  companies.   Through  creative  collaborations  with
technology  leaders,  we believe  we can  accelerate  and  expand  our  internal
discovery  efforts.  We have  entered  into a number  of  alliances  with  other
platform  technology  companies to enhance our drug  discovery  and  development
capabilities. These alliances include:

o        our alliance with Rigel, Inc. to use Rigel's intellectual  property and
         expertise to discover novel protein targets involved in neural cell and
         antibody activation;




o        our alliance with Arena  Pharmaceuticals  involving the  application of
         Arena's constitutive activation technology to a family of receptors;

o        our  alliance  with Array  Biopharma,  Inc. to design and  synthesize a
         focused library of small molecules; and

o        our  alliance  with Caliper  Technologies  Corp.  to utilize  Caliper's
         proprietary microfluidics technology to screen against our targets.

            Outsource Capital Intensive and Non-Strategic  Activities. We intend
to focus our resources on research and development activities by outsourcing our
requirements  for  clinical  drug  supply and  certain  preclinical  studies and
clinical monitoring activities.  We believe the availability of skilled contract
manufacturers and contractors will allow us to cost-effectively meet these needs
and thereby allow us to concentrate our full attention and resources on our core
discovery and development programs to generate additional product opportunities.


Our Product Pipeline

            The following table summarizes our most advanced product  candidates
currently  in  preclinical  or  clinical  development  and  those  currently  in
research, and is followed by detailed descriptions of each program:




- -------------------------------------------------------------------------------------------------------
Program                   Compound       Targeted Indication      Status        Commercial Rights
- -------------------------------------------------------------------------------------------------------
                                                                    
PRODUCTS UNDER DEVELOPMENT:

GABA-A Agonist            NBI-34060      Insomnia                Phase II       Neurocrine

CRF R1 Antagonist         NBI-37582      Anxiety, Depression     Preclinical    Janssen/ Neurocrine

CRF R1 Antagonist         NBI-34041      Anxiety, Depression     Phase I        Neurocrine

IL-4 Fusion Toxin         NBI-3001       Malignant Glioma        Phase II       Neurocrine

IL-4 Fusion Toxin         NBI-3001       Additional Cancers      Preclinical    Neurocrine
                                         (kidney, lung)
Altered Peptide Ligand    NBI-5788       Multiple Sclerosis      Phase II       Neurocrine

Altered Peptide Ligand    NBI-6024       Type 1 Diabetes         Phase I/II     Taisho/Neurocrine

GnRH Antagonist                          Endometriosis,          Preclinical    Neurocrine
                                         Prostate Cancer
RESEARCH:

Excitatory Amino Acid Transporters       Neurodegenerative       Research       Wyeth-Ayerst/
                                         Diseases                               Neurocrine
CRF R1 Antagonist                        Gastrointestinal        Research       Neurocrine
                                         Disorders
CRF R2 Antagonist                        Eating Disorders        Research       Neurocrine

Urocortin/CRF R2 Agonist                 Obesity                 Research       Eli Lilly

Melanocortin Receptor Agonist            Obesity                 Research       Neurocrine

Melanin Concentrating Hormone            Obesity                 Research       Neurocrine
Antagonist

Hypocretin Agonist/Antagonist            Sleep Disorders         Research       Neurocrine
- --------------------



<FN>

     "Phase II" indicates that we, or our collaborators, are conducting clinical
trials on groups of  patients  afflicted  with a  specific  disease  in order to
determine preliminary efficacy, optimal dosages and expanded evidence of safety.

       "Phase  I"  indicates  that  we,  or our  collaborators,  are  conducting
clinical trials to determine early safety profile,  maximally tolerated dose and
pharmacological properties of the product in human volunteers.

     "Preclinical"  indicates  that a drug candidate is being  selected,  or has
been selected,  and is undergoing  toxicology studies and manufacturing to allow
for Phase I clinical trials.

     "Development"  indicates that lead compounds have been discovered that meet
certain  laboratory  and  preclinical  criteria.  These  compounds  may  undergo
structural  modification  and more extensive  evaluation  prior to selection for
preclinical development.

     "Research"   indicates   identification  and  evaluation  of  compounds  in
laboratory and preclinical models.
</FN>


Products Under Development

     GABA-A Agonist

         Insomnia is a  prevalent  neurological  disorder in the United  States,
with  up to 58% of the  adult  population  reporting  one or  more  symptoms  of
insomnia  a few  nights  per  week or  more,  according  to the  National  Sleep
Foundation. Despite this widespread prevalence, insomnia remains a disorder with
high unmet medical needs, including the ability to maintain sleep throughout the
night without next-day  residual  effects.  Researchers have found that insomnia
can be treated by drugs that interact with the site of action of a natural brain
chemical  involved in promoting and maintaining  sleep.  This chemical is called
gamma  amino-butyric  acid, or GABA, and the site of action is called the GABA-A
receptor.

         During the 1980s,  a class of drugs that  targets the GABA-A  receptor,
known as  benzodiazepines,  was used as  sedatives to treat  insomnia.  The most
well-known of the  benzodiazepines  is Valium(R).  This class of drugs  produced
several  undesirable side effects,  including  negative  interactions with other
central  nervous  system  depressants,  such  as  alcohol,  the  development  of
tolerance  upon repeat dosing,  insomnia  following  discontinuation  of dosing,
hangover effects the next day, and impairment of coordination and memory. Memory
impairment,  which can include  amnesia for events  occurring prior to and after
drug  administration,  is of particular  concern in the elderly whose  cognitive
function may already be impaired by the aging process.  During the late 1980s, a
class of drugs  targeting  a  specific  site on the  GABA-A  receptor,  known as
non-benzodiazepines,  was  developed.  The  non-benzodiazepines  reduce the side
effects   associated   with   benzodiazepines.   The   most   popular   of   the
non-benzodiazepines  are marketed as Ambien(R) and  Sonata(R).  Ambien(R) is the
current leader with worldwide sales in 2000 of approximately $840 million.

         Our  drug  candidate  for  the  treatment  of  insomnia,  NBI-34060,  a
non-benzodiazepine,  acts on a  specific  site  on the  GABA-A  receptor.  It is
through  this   mechanism   that  the  currently   marketed   non-benzodiazepine
therapeutics also produces their sleep-promoting effects. However,  NBI-34060 is
more potent than the currently marketed non-benzodiazepines, including Ambien(R)
and Sonata(R),  and is more selective than the  benzodiazepines for the specific
subtype of receptors  within the brain believed to be responsible  for promoting
sleep.  We believe that this improved  profile and more selective drug targeting
will reduce the side effects  characteristic of the currently marketed products.
We also  believe  that  receptor  binding  studies  and  preclinical  studies on
NBI-34060  indicate  that it is a highly potent GABA-A  receptor  activator,  or
agonist,  that acts very  specifically on the receptor subtype we are targeting.
NBI-34060  also appears to be devoid of next day hangover  effects and we expect
it to have a considerably  reduced amnesic  potential.  The elderly  population,
which  represents  a  large  portion  of  the  insomnia  market,  would  benefit
especially from a novel therapeutic with an improved safety profile, rapidity of
onset and decrease in memory impairment.

         Researchers  designed the first Phase I clinical  trial on NBI-34060 to
determine  the safety and  tolerance  of  NBI-34060  and  provide a  preliminary



evaluation  of the sedative  potential in 42 normal  volunteers  as reflected in
self-ratings of drowsiness, disruption of memory and impairment of coordination.
In this trial,  subjects  tolerated  NBI-34060  well, and reported no serious or
unexpected adverse side effects. The subjects  consistently reported drowsiness,
indicating  strong  potential  for  the  sedative  properties  of the  compound.
Subsequently,  in the first  quarter  of 1999,  we  completed  a second  Phase I
clinical  trial in 30  healthy  volunteers  to  further  explore  the safety and
kinetic  profile  of  NBI-34060.  As  demonstrated  in the first  Phase I trial,
NBI-34060 demonstrated a very good safety profile.

         In  1999,  we  completed  a Phase  II  placebo-controlled  multi-center
clinical  trial  evaluating  the  efficacy of  NBI-34060  in 228  subjects  with
transient insomnia.  Transient insomnia means occasional sleeplessness caused by
environmental  factors,  such as  jetlag.  The  trial was  conducted  in a sleep
laboratory  employing  objective  assessments  of sleep  onset and  safety.  The
results  indicated that NBI-34060 is safe and effective in helping subjects with
transient  insomnia  achieve rapid sleep  without the next day residual  effects
associated with most currently marketed  sedatives.  The results showed that the
primary  clinical  endpoint and the required  regulatory  endpoint for approval,
time to sleep onset, was reached at a statistically  significant  level. In this
trial,  those  subjects  receiving  NBI-34060  took a mean time of 16 minutes to
progress to sleep onset  versus a mean time of 34 minutes in the placebo  group.
These  results  were  statistically  significant  at p < 0.001.  This means that
applying widely used  statistical  methods,  the chance that these results could
have  occurred by accident  was less than one in 1,000.  In  addition,  the data
indicated  that a majority of subjects in the treated  group fell asleep  within
9.5  minutes as  indicated  by the median  time to sleep onset as compared to 23
minutes in the placebo group.

         Results of a Phase II clinical trial comparing NBI-34060, Ambien(R) and
zopiclone  relative to placebo  during middle of the night dosing,  demonstrated
that NBI-34060 does not lead to next-day hangover effects,  while both Ambien(R)
and zopiclone exhibited  statistically  significant measures of next-day adverse
side effects of residual sedation. In two clinical trials evaluating the effects
of NBI-34060 on the largest  target  populations  for insomnia  (the elderly and
females),   results   demonstrated   that   NBI-34060   showed  no   significant
pharmacokinetic  differences  in age or gender.  In a randomized,  double-blind,
placebo-controlled  clinical study with multiple doses of NBI-34060 conducted in
young adults and elderly subjects, results demonstrate that NBI-34060 works as a
sedative-hypnotic  with no major differences in the pharmacokinetics for maximum
plasma levels or total drug exposure between young adults and elderly  subjects.
Also,   there  were  no  changes  between  these  patient   populations  in  the
accumulation  of  NBI-34060  after  four  consecutive   nightly  doses.   Safety
evaluation and subjective  measures of next-day  residual effect  confirmed that
the drug was well-tolerated in both groups with the expected sedation during the
night, and with no hangover or residual carry over next-day effect.

         Based on the results from our Phase II trials,  we have moved to expand
clinical  development of NBI-34060.  In 2000 we initiated  nine clinical  trials
involving 503 subjects to evaluate various  formulations and patient  subgroups.
Among  other  research  goals,  we  intend to  determine  whether  NBI-34060  is
effective in treating chronic  insomnia,  which is  sleeplessness  not caused by
environmental  factors.  We are  developing a formulation  of NBI-34060 to treat
chronic insomnia. We are also designing a large-scale pivotal Phase III program,
from  which we expect to  determine  the  approvability  of the drug  candidate.
Currently, we expect to begin this pivotal trial in the second half of 2001.

         Another   important  feature  of  NBI-34060  is  its  relatively  short
half-life,  or duration of action of the  compound,  in the body.  The levels of
NBI-34060  in the blood  stream  reach the  highest  point 30 minutes  after the
subject takes the tablet.  The NBI-34060 is then rapidly  removed from the blood
stream so it cannot be  detected  four  hours  later.  This  rapid  peak of drug
results in rapid  sleep  onset  followed  by rapid  removal of the drug from the
body,  reducing the risk of next-day  effects of the drug.  We believe that this
short  duration  of action  will  allow for  bedtime  dosing for people who have
trouble falling asleep and dosing in the middle of the night for people who have
trouble  staying asleep  without  causing the side effects and next day hangover
that occurs with the longer acting drugs like Ambien(R) and the benzodiazepines.
We also  believe  that this short  duration of action will allow us to formulate
the drug in a modified release form that will  effectively  provide two doses of
drug,  a bedtime  dose and a middle of the night dose,  which will both  rapidly
induce sleep and maintain  sleep through the night.  If  successful,  this would
represent  the first  non-benzodiazepine  approved  by the FDA for  maintaining,
rather than simply inducing, sleep.

         We face  the risk  that  the  side  effects  and  efficacy  profile  of
NBI-34060  seen in our Phase I and II trials may not be confirmed in  additional
clinical  trials or that the results of future  trials may not  warrant  further
trials.





     Corticotropin-Releasing Factor

         According to the Surgeon  General's 1999 Report on Mental Health,  6.5%
of the U.S. adult population  experiences a major  depressive  episode each year
and  16.4%  of the U.S.  adult  population  has an  anxiety  disorder.  Existing
anti-depressant  and anti-anxiety  therapeutics sold approximately $11.1 billion
worldwide in 1999,  with sales  anticipated  in excess of $12.6  billion in 2000
according to market  analyst  reports from  Datamonitor.  However,  there remain
significant unmet medical needs. The leading drug class,  known as the selective
serotonin re-uptake inhibitors, is not effective in one-third of patients. These
drugs  frequently  require  as long as  three  weeks  to take  effect,  and have
significant adverse side effects such as sexual dysfunction.  Therefore, a rapid
acting  anti-depressant  with fewer side effects would represent a major advance
in the treatment of depression.  Corticotropin-releasing  factor antagonists may
provide such an advance in anti-depressant  therapy through a specific mechanism
for combating the hormonal abnormalities associated with depression.

         Researchers  have  identified  what  they  believe  to be  the  central
mediator  of the body's  stress  responses  or  stress-induced  disorders.  This
mediator  is  a  brain   chemical  known  as   corticotropin-releasing   factor.
Corticotropin-releasing  factor is overproduced in clinically depressed patients
and  individuals  with  anxiety  disorders.   Current  research  indicates  that
clinically  depressed patients and patients with anxiety experience  dysfunction
of the  hypothalamic-  pituitary-adrenal  axis, which is the system that manages
the  body's  overall  response  to  stress.  When the body  detects  a threat to
physical  or  psychological  well-being,  the  region  of the brain  called  the
hypothalamus  amplifies  production  of  corticotropin-releasing  factor,  which
induces the physical  effects that are associated with stress and which can lead
to depression or anxiety.

         The  novelty  and  specificity  of the  corticotropin-releasing  factor
mechanism  of action and the  prospect of  improving  upon  selective  serotonin
re-uptake  inhibitor  therapy is a topic of interest  throughout the psychiatric
community and pharmaceutical industry, representing a market opportunity both to
better serve  patients and expand  overall  treatment for this life  threatening
disease.

         We have a strategic position in the corticotropin-releasing  factor, or
CRF, field through our intellectual  property  portfolio and  relationship  with
experts in the  neuropsychiatric  field.  Wylie Vale,  Ph.D., our co-founder and
Chief  Scientific  Advisor,  is  considered  to be a  leader  in this  field  of
research.  We have  further  characterized  the CRF  receptor  system  and  have
identified  additional  members of the CRF  receptor  family.  We have  received
patents on two receptor  subtypes  called CRF R1 and CRF R2, and we have pending
patent  applications  on small  molecule  organic  compounds  modulating the CRF
receptors.

         Depression.  Depression is one of a group of neuropsychiatric disorders
that is characterized  by extreme feelings of elation and despair,  loss of body
weight,  decreased  aggressiveness  and  sexual  behavior,  and  loss of  sleep.
Researchers  believe that depression results from a combination of environmental
factors, including stress, as well as an individual's biochemical vulnerability,
which is genetically  predetermined.  Researchers  believe that the  biochemical
basis of depression involves elevated secretion of CRF and abnormally low levels
of other  neurotransmitters in the brain such as serotonin.  The most frequently
prescribed  antidepressant therapies are selective serotonin reuptake inhibitors
such as Prozac(R),  Zoloft(R),  Paxil(R) and Celexa(R) which act to increase the
levels of serotonin and several other chemicals in the brain.  However,  because
these drugs affect a wide range of neurotransmitters,  they have been associated
with a number of adverse side effects.  While newer,  more selective drugs offer
some safety improvement,  side effects remain problematic.  In addition,  one of
the biggest limitations of most existing anti-depressant therapies is their slow
onset of action.

         In our CRF R1 antagonist program, our corporate  collaborator,  Janssen
Pharmaceutica,  selected a CRF R1 receptor antagonist drug candidate, NBI-30775,
for  preclinical  studies in 1996.  Janssen  initiated and completed a number of
Phase I clinical  trials on the compound in late 1998 and  initiated a Phase IIa
open label trial in 1999.  Results from this trial  indicated that NBI-30775 was
safe and well-tolerated and demonstrated anti-depressant activity as measured by
a widely-accepted  depression scale known as the Hamilton  Depression Scores. In
this trial, Janssen administered  NBI-30775 to 20 patients with major depressive
disorders.  Results  from the trial,  as reported in the Journal of  Psychiatric
Research,  showed  that  treatment  response,  as  defined  by  more  than a 50%
reduction in Hamilton Depression Scores,  occurred in 50% of the patients in the
low dose  group  and 80% of the  patients  in the  higher  dose  group.  We were
strongly encouraged by these results,  which we believe support the hypothesized
mechanism of action.  While this trial found  NBI-30775  to be safe,  reversible
increases in liver  enzymes  occurred in two  volunteers  in an expanded  safety
study. As a result, Janssen announced its decision to discontinue development of



NBI-30775.  However,  because of the positive  efficacy  results for  NBI-30775,
Janssen decided to proceed with a back-up compound  identified from its research
relationship with us. In 2000,  Janssen selected a back-up candidate to develop,
NBI-37582. This candidate is currently in late preclinical testing.

         In addition to our CRF R1 program with  Janssen,  we are  conducting an
independent  CRF R1 antagonist  program  focused on a series of our  proprietary
chemical  compounds.  We have selected  several lead candidates for this program
and  entered  into  Phase I  clinical  trials in  December  2000.  This Phase I,
randomized, double-blind, placebo-controlled, single-dose trial was conducted in
48  normal  healthy   volunteers  and  was  designed  to  evaluate  the  safety,
tolerability,   pharmacokinetics,  and  pharmacodynamics,   including  endocrine
profiles,  over  a  range  of  six  escalating  doses.  Initial  pharmacokinetic
evaluation indicates rapid absorption and good  dose-proportionality  and plasma
half lives  supportive of a once-a-day  dosing  schedule.  No safety issues were
noted  which  would  preclude  advancement  into  the  next  phase  of  clinical
evaluation. A two week multiple dose, dose-escalating Phase I placebo controlled
clinical  trial will be initiated to further  evaluate the safety and  endocrine
profiles  of this  compound  to prepare  for Phase II  efficacy  evaluation.  We
anticipate initiating Phase II clinical trials in the second half of 2001.

         We face the risk that CRF R1 antagonist  compounds may not be effective
and safe  therapeutics for the treatment of depression or any other  conditions.
In addition,  Janssen or we may decide not to initiate Phase I clinical  testing
or progress to later clinical trials in a timely manner, if at all.

         Anxiety.  Anxiety is among the most commonly  observed group of central
nervous system  disorders,  which includes  phobias or irrational  fears,  panic
attacks,  obsessive-compulsive  disorders and other fear and tension  syndromes.
The  Surgeon  General's  1999 Report on Mental  Health  estimates  that  anxiety
disorders  affect  16.4% of the U.S.  adult  population.  Of the  pharmaceutical
agents  that  other  companies  currently  market for the  treatment  of anxiety
disorders,  benzodiazepines,  such as  Valium(R)  and  Xanax(R),  are  the  most
frequently prescribed. Several side effects, however, limit the utility of these
anti-anxiety  drugs. Most problematic among these are drowsiness,  the inability
to stand up,  amnesia,  drug dependency and withdrawal  reactions  following the
termination  of  therapy.   Despite  these  adverse  effects,   total  sales  of
benzodiazepines  were approximately $800 million in 1999. In view of significant
evidence implicating CRF in anxiety-related  disorders,  we are developing small
molecule  CRF R1  receptor  antagonists  as  anti-anxiety  agents that block the
effects of  overproduction  of CRF. We believe  that these  compounds  utilize a
novel  mechanism of action that may offer the advantage of being more selective,
thereby  providing  increased  efficacy with reduced side effects as compared to
benzodiazepines.

         With our corporate collaborator, Janssen, and in our independent CRF R1
antagonist program,  we are developing small molecule  therapeutics to block the
effects of  overproduction of CRF in anxiety.  As a co-examined  variable in the
Janssen  open label Phase IIa clinical  trial for  depression  described  above,
Janssen  analyzed  the  anti-anxiety  effects of the CRF R1 receptor  antagonist
NBI-30775  using the Hamilton  Anxiety Scores.  Janssen  observed a reduction in
Hamilton  Anxiety  Scores from  baseline in both  treatment  groups at all times
after dosing. In addition,  in preclinical studies used to evaluate anti-anxiety
drugs,  our scientists have  demonstrated  with  statistical  significance  that
clinical compound  candidates from our independent CRF R1 antagonist program are
effective  following  oral  administration.  We did not observe any  evidence of
clinically  relevant side effects.  These results are  encouraging  because they
suggest that compounds blocking the CRF R1 receptor may be effective in treating
anxiety-related disorders. Despite these early results, Janssen or we may decide
not to initiate  clinical  testing of CRF R1  antagonist  compounds for anxiety.
Even if those trials are conducted, the data may not support continuation of the
program and additional clinical trials.


     IL-4 Fusion Toxin

         Interleukin  4, or IL-4, is a natural  chemical,  which  modulates cell
growth.  Proteins  that  bind to  IL-4,  known  as IL-4  receptors,  are  highly
concentrated  on the  cells of  malignant  brain  tumors  as well as many  other
cancers,  including some types of kidney and lung cancers. Targeted toxins are a
novel form of anti-cancer  therapy under  investigation in a variety of clinical
settings.  Targeted toxin  therapeutics carry a bacterial toxin to a target site
on the cancer  cells and  subsequently  kills the cancer cell.  Many  scientists
believe that targeted toxins have several potential advantages over conventional
chemotherapy  in that they are more  selective and effective in the treatment of
chemotherapy-resistant cancer cells.



         In 1998, we exclusively licensed from the National Institutes of Health
a  targeted  toxin  compound,  IL-4  fusion  toxin,  which we call  NBI-3001.  A
collaboration  between the FDA and the National  Cancer  Institute  designed the
IL-4  fusion  toxin.  It is a  combination  protein in which IL-4 is attached to
Pseudomonas exotoxin, a bacterial toxin that can kill cells. The IL-4 portion of
the fusion toxin  preferentially  binds to human cancer cells because the cancer
cells express elevated levels of receptors for IL-4 on their surface, while IL-4
receptor  expression is absent or undetectable  in normal tissue.  Once the IL-4
portion of the IL-4  fusion  toxin  targets the toxin to the cancer  cells,  the
toxin portion of the molecule preferentially kills the cancer cells.

         Malignant  Glioma.  Malignant  brain tumors are a significant  cause of
cancer  death.  The most  common form of  malignant  brain  cancer is  malignant
glioma. These tumors arise within the brain and generally remain confined to the
brain. According to the American Cancer Society, despite surgical, radiation and
chemotherapy  treatments,  the median survival rate for malignant glioma is only
in the range of 9 to 12  months.  The  clinical  course of  malignant  glioma is
characterized  by  relentless  loss of vital  neurological  functions  and death
within approximately 12 months.

         In 1999 we  initiated a Phase I/II trial of  NBI-3001 in patients  with
malignant  glioma  in  which  the  primary   endpoints  were  safety  and  tumor
regression.  We  completed  this trial in June 2000.  We  enrolled a total of 31
patients  with  recurrent  gliomas,  which  were  unresponsive  to  surgery  and
radiotherapy in the trial. Our researchers  treated  patients with  intratumoral
infusions of NBI-3001 for up to four days.  This trial found NBI-3001 to be safe
and to have an acceptable  degree of  tolerability  in this patient  population.
While  approximately  one-third of the patients exhibited side effects during or
immediately  following therapy,  these effects were consistent with marked tumor
cell death and the  subsequent  inflammatory  response to this tumor cell death.
The  researchers  did  not  observe  any  significant  peripheral   drug-related
toxicities.  The  researchers  reported  that,  of the 27 patients who completed
therapy:

o        seven patients, or 26%, experienced complete remissions,  defined as no
         evidence of viable tumor;

o        10 patients, or 37%, experienced a partial response, defined as greater
         than 50% reduction in tumor mass; and

o        10 patients,  or 37%,  continued  to suffer from stable or  progressive
         disease.

     In addition,  the  six-month  median  survival  data showed  trends  toward
efficacy.  We initiated  an  additional  confirmatory  Phase II trial to further
establish dosing regiment, safety and efficacy in the fourth quarter of 2000. We
also plan to initiate a Phase III trial in the second half of 2001.

     In October 1999,  the FDA granted us fast track  designation  for NBI-3001.
Fast track designation allows us to accelerate our clinical program for NBI-3001
and expedite  receipt of  regulatory  approvals.  In April 2000, we were awarded
orphan drug  designation  for NBI-3001 for astrocytic  glioma.  Under FDA rules,
drug  developers  may  obtain  orphan  drug  designation  for drugs that treat a
disease or condition that affects fewer than 200,000 people in the United States
per year.  Orphan drug  designation  provides  us with seven years of  marketing
exclusivity  following approval,  tax incentives and access to grant funding. We
face the  risk  that we will  not  successfully  complete  clinical  testing  or
progress to later clinical trials in a timely manner, or at all.

     Additional  Cancers. In conjunction with our clinical trials of IL-4 fusion
toxin  in  malignant  glioma,  we  entered  into a  collaborative  research  and
development  agreement  with the FDA to  investigate  the safety and efficacy of
IL-4 fusion toxin in  laboratory  models of  different  cancers and by different
routes of  administration.  Research  teams from the FDA and NIH have  published
results  with IL-4  fusion  toxin  demonstrating  a high  level of  binding  and
destruction of specific types of cancers. We are conducting preclinical research
to support the application of NBI-3001 to peripheral solid tumors and have shown
that IL-4 fusion toxin can be safely  administered  intravenously in preclinical
models.  We plan to initiate a Phase I clinical  trial in the second  quarter of
2001 to first investigate the safety and efficacy of NBI-3001 against kidney and
non-small-cell  lung  cancers.  We face  the  risks  that the  effectiveness  of
NBI-3001 seen in our laboratory  models,  or the safety profile of NBI-3001 seen
in our preclinical  models,  may not be confirmed in clinical trials or that the
results of future clinical trials may not warrant further  development in any of
these  settings or that the trial  results may not support  initiating  clinical
trials in cancers other than malignant glioma.





     Altered Peptide Ligands

         The American  Autoimmune  Related Diseases  Association  estimates that
over 50 million people in the United States suffer from autoimmune diseases such
as multiple sclerosis,  rheumatoid  arthritis,  Type 1 diabetes,  systemic lupus
erythematosus and thyroiditis.  Scientists believe that the body's immune system
causes these  diseases.  The immune system  protects an individual  from disease
agents,  such as viruses, by recognizing and destroying those foreign substances
using  specialized  blood  cells,  called  lymphocytes.  Occasionally,  however,
certain lymphocytes arise that inappropriately  recognize the body's own tissues
as an  invader  and begin to  attack  normal  healthy  tissue,  resulting  in an
autoimmune  disease like Type 1 diabetes.  While the mechanism through which the
lymphocyte initiates the autoimmune disease is still unknown, the development of
drugs that  specifically  suppress the action of  self-reactive  lymphocytes  or
restore  the  function  of  regulatory  cells  may  prove  advantageous  for the
prevention, cure or treatment of an autoimmune disease.

         One type of  lymphocyte  involved  in the  immune  response  to self or
foreign  substances  is the T cell. T cells detect  antigens,  which are foreign
substances,  such as viruses,  bacterias or other proteins the T cell recognizes
as foreign.  T cells  recognize  these  antigens and destroy  them.  Experiments
conducted by our scientists  determined that specific amino acid residues within
a peptide  sequence  of an  antigen  are  required  for T cell  recognition  and
subsequent cellular activation.  Scientists can specifically alter the structure
of such a peptide  fragment so that it will not  properly  activate  the T cell.
This  altered  peptide  ligand can thus prevent the T cell from  destroying  its
target,  or in the  case of an  autoimmune  reaction,  prevent  the T cell  from
destroying the healthy tissue.

         Multiple Sclerosis.  Multiple sclerosis is a chronic autoimmune disease
characterized  by recurrent  attacks of neurologic  dysfunction due to damage in
the central nervous system.  The classic clinical features of multiple sclerosis
include impaired vision and weakness or paralysis of one or more limbs. Patients
develop a slow,  steady  deterioration  of  neurologic  function over an average
duration of approximately 30 years. According to the National Multiple Sclerosis
Society,  there are an  estimated  350,000  cases of multiple  sclerosis  in the
United  States and a similar  number of patients  in Europe  with  approximately
17,000 new cases diagnosed worldwide each year.  Currently available  treatments
for multiple  sclerosis  offer only limited  efficacy.  Doctors often  prescribe
steroids  to  reduce  the  severity  of  acute  flare-ups  and  speed  recovery.
Experimental  therapy  with other  immune-suppressive  agents has shown  limited
success.

            Our  co-founder,  Dr.  Lawrence  Steinman,  identified  one  of  the
dominant  destructive  T cell  types in the brains of  patients  who had died of
multiple sclerosis. Dr. Steinman further identified one of the dominant antigens
on the normal cell targeted by the  autoreactive T cells, a peptide from a brain
protein know as myelin basic protein. We designed a novel altered peptide ligand
based on myelin basic  protein that would target the  autoreactive  T cells that
cause  neurological  damage  in  multiple  sclerosis.   Together  with  Novartis
Pharmaceuticals  Corporation, our former collaborative partner for this program,
we  filed an  investigational  new drug  application  with the FDA and  received
approval in 1996 to commence clinical trials. We subsequently  completed Phase I
clinical  trials,  and  initiated  two Phase II  clinical  trials of our altered
peptide ligand for the treatment of multiple sclerosis, NBI-5788, in 1999.

         The  first  Phase  II trial  was a  multi-center,  placebo  controlled,
randomized  parallel group design  involving  three doses of the altered peptide
ligand in 5, 20, and 50 mg weekly  doses for four  months in 13 centers in North
America  and  Europe.  In July 1999  while the  Phase II trials  were  underway,
Novartis  exercised its right to terminate the  collaboration  effective January
2000.  Subsequently,  the  Data  and  Safety  Monitoring  Board  for  the  trial
recommended,   and  we  agreed,   that  administration  of  the  trial  drug  be
discontinued  based on reports of adverse  allergic  reactions.  We continued to
evaluate  all of the enrolled  patients in the study  through  December  1999 in
accordance with the study protocol. We reacquired all rights to the program from
Novartis on January 7, 2000 and initiated data analysis. The final data analysis
from the multi-center  trial showed no increases in either clinical  relapses or
in new  lesions in all  patients,  even those with  allergic  reactions.  Of the
patients  completing the  double-blind  phase of the study,  the total volume of
enhancing  lesions  was  reduced  in  the  5  mg  dose  group  compared  to  the
placebo-control patents (p<0.029 Mann Whitney for two treatments).  We could not
conduct this same secondary  analysis in the 20 mg or 50 mg groups,  since there
were not enough  patients with positive  scans for us to evaluate their magnetic
resonance  imaging  changes.  Moreover,  57% of the  patients  in the 5 mg group
experienced reductions in the volume of new enhancing lesions compared to 25% in
the placebo group.  Because of these factors, we believe that for this compound,
optimal dosing may be at lower levels, and we are currently planning a Phase IIb
trial to  establish  the  efficacy  profile  and  optimum  dosing  regiment  for
NBI-5788.




         The second Phase II trial, which we conducted in collaboration with the
National Institutes of Health,  involved an open-label,  unblinded,  non-placebo
control trial in eight patients,  seven of whom received multiple  injections of
50 mg weekly while the final subject received 5 mg. In this trial,  published in
Nature Medicine, the authors observed a higher incidence of new brain lesions in
two  patients  who  received 50 mg doses and the one  patient who  received 5 mg
doses. As a result, the trial was stopped.  However, the authors did not provide
direct evidence that NBI-5788 triggered the lesions.

         Our aim for future  trials will be to further  establish the benefit of
low-dose altered peptide ligand therapy in patients with multiple sclerosis.  We
face the risks that we may not initiate or complete  additional  clinical trials
or that  results  of any  such  studies  may  not  warrant  additional  clinical
development of potential products.

         Type 1 Diabetes. Utilizing our experience in the development of altered
peptide ligands for multiple sclerosis, we have extended this approach to Type 1
or juvenile-onset  diabetes, which is a metabolic disease in which the body does
not produce  enough  insulin.  Like multiple  sclerosis,  in Type 1 diabetes the
immune system  erroneously  targets healthy tissue,  in this case the pancreatic
cells  responsible for the production of insulin.  Type 1 diabetes is one of the
most  prevalent  chronic  childhood  conditions  in  North  America,  afflicting
approximately one million patients in 1999. Diabetics often suffer from a number
of complications of the disease including heart disease,  circulatory  problems,
kidney failure,  neurologic disorders and blindness.  Current therapy for Type 1
diabetes consists of daily insulin injections to regulate blood glucose levels.

          We believe that an altered  peptide  ligand  specific for autoimmune T
cells  involved in diabetes may stop the  destruction  of the  insulin-secreting
cells in  pre-diabetic  patients,  thus  allowing them to delay or avoid chronic
insulin therapy. Working with leading diabetologists at the Barbara Davis Center
for  Childhood  Diabetes at the  University  of Colorado,  our  scientists  have
engineered an altered  peptide  ligand that affects  immune cells  targeting the
pancreas.  In preclinical studies,  this altered peptide ligand,  NBI-6024,  was
capable of eliciting a protective  immune response and reducing the incidence of
diabetes. In addition,  experiments using immune cells derived from the blood of
Type  1  diabetes  patients  indicate  that  patients'  immune  cells  recognize
NBI-6024. This suggests that NBI-6024 may have the potential to intervene in the
disease  process  in  humans.  We  have  completed  2 Phase I  safety  and  dose
escalating clinical program in diabetic patients.  These trials included 50 Type
1 diabetic  patients.  Data from this trial  indicates that NBI-6024 is safe and
well tolerated.  We are enrolling  patients for an additional Phase I multi-dose
trial and expect to  initiate  the first  Phase II trial in early 2001 to assess
the safety and  biological  activity  of  multiple  doses of  NBI-6024 in adult,
adolescent and pediatric  patients with Type 1 diabetes and a Phase II/III trial
in the third quarter of 2001.

         In  January   2000,   we  entered   into  an   agreement   with  Taisho
Pharmaceutical  Co., Ltd.  providing  Taisho with an exclusive  option to obtain
European,  Asian and North  American  rights to NBI-6024.  In July 2000,  Taisho
exercised  the option as to European  and Asian  rights,  and we granted  Taisho
exclusive  rights  to  NBI-6024  in  those  regions.   Under  the  collaboration
agreement,  we will  receive  licensing  and option  fees,  payments for certain
development and regulatory  milestones,  significant  reimbursement of worldwide
development  expenses and  payments  based on sales upon  commercialization.  In
November,  2000 we expanded our collaboration  with Taisho,  granting Taisho the
exclusive  rights to develop and  commercialize  NBI-6024  in North  America and
other  countries  outside of Europe and Asia.  The  worldwide  collaboration  is
valued at up to $100 million,  including all potential  licensing fees, purchase
fees, milestones and development expenses.

         We  face  the  risk  that  large-scale  studies  of our  drug in Type 1
diabetes  patients may show different  results than our  preclinical  studies in
animals  and in cells  derived  from  Type 1  diabetes  patients  or our Phase I
trials.


     Gonadotropin-Releasing Hormone Receptor

         Gonadotropin-releasing  hormone,  or  GnRH,  is a  brain  peptide  that
stimulates the secretion of the pituitary  hormones that are responsible for sex
steroid production and normal reproductive function. Researchers have found that
chronic  administration of GnRH agonists  reversibly shuts down this transmitter
pathway and is clinically useful in treating  hormone-dependent diseases such as
prostate  cancer and  endometriosis.  Other  companies  have  developed  several
peptide drugs on this principle, such as Lupron(R) and Zoladex(R), and according
to market analyst reports by Euromonitor and Epicom Business Intelligence, these
drugs now have an  estimated  market  size in excess  of $2.0  billion  annually
worldwide. However, since these drugs are peptides, they must be injected rather



than taken  orally.  These types of agonists  take up to several  weeks to exert
their  effect,  a factor  not seen with  direct  gonadotropin-releasing  hormone
antagonists.  Until these drugs exert their effects,  they have shown a tendency
to exacerbate the condition.

         We  believe  that  there  is a large  market  potential  for an  orally
delivered  gonadotropin-releasing  hormone  antagonist  that  does  not have the
tendency to initially exacerbate the patient's  condition.  We have screened our
small molecule library and conducted structure activity studies to produce small
molecule  orally-active  gonadotropin-releasing  hormone  antagonists.  We  have
identified  several  series  of  small  molecule  compounds  and are  conducting
additional studies to select a final clinical  candidate.  We intend to select a
lead clinical candidate in the first half of 2001 and expect to initiate Phase I
clinical trials in the second half of 2001. We believe that the results of these
Phase I studies will be predictive of efficacy in many potential indications. We
face the risk that our work in this area may not lead to clinical  candidates or
that, even if we select a candidate, clinical trials may show it is not safe and
effective.

         We plan to focus our  clinical  efforts on  prostate  cancer and in the
area  of  women's  health,   including   endometriosis,   uterine  fibroids  and
infertility.  According to a 1993 article in  Contemporary  OB/GYN,  researchers
believe  that more than five  million  women in the U.S.  alone are  affected by
chronic endometriosis, representing approximately 10% prevalence in reproductive
age women. Of those afflicted,  approximately  200,000 patients are treated in a
hospital  setting,  and an  additional  250,000  are  treated  in an  outpatient
setting, according to a 1998 National Patient Profile audit. We believe that the
availability  of an oral  treatment  lacking  the  side  effect  profile  of the
currently  available  peptide  agonists  may be an  alternative  to surgery  and
encourage a higher treatment rate. We also believe our drug will have utility in
the treatment of prostate  cancer,  which has over 180,000 new cases per year in
the U.S., according to the American Cancer Society.


Research

     Excitatory Amino Acid Transporters

         Some of the most  successful  central  nervous system drugs,  including
selective  serotonin reuptake  inhibitors such as Prozac(R),  selectively target
brain amino acid  transporters.  Similarly,  we are  targeting a set of proteins
generally  located in the brain which  transport  brain  chemicals in and out of
cells,  called excitatory amino acid  transporters,  to selectively  control the
levels of a brain  chemical  called  glutamate in order to produce a therapeutic
benefit in disorders  where  glutamate  levels are abnormal such as head trauma,
schizophrenia,  Lou Gehrig's Disease and other neurodegenerative and psychiatric
disorders.

         We are  collaborating  with  Wyeth-Ayerst  Laboratories,  a division of
American Home Products Corporation, to control glutamate transporter function as
a  novel  strategy  for  the  treatment  of  neurodegenerative  and  psychiatric
disorders.  Our collaboration includes basic research to understand the function
and  regulation  of  the  transporters,   along  with  the   identification  and
characterization of chemical and biological leads. We face the risks that we may
be unable to  demonstrate  that these  excitatory  amino acid  transporters  are
therapeutic  targets or that we may fail to identify any product  candidates for
preclinical or subsequent clinical development.

         In 2000 we expanded our excitatory amino acid transporter  research and
initiated  a research  program  focused on retinal  cell death  associated  with
damage from low blood  flow.  The  National  Institutes  of Health  awarded us a
research grant to fund our work to identify novel  compounds for the alleviation
of  neuronal  cell death in  response  to a wide range of  conditions  including
diabetic induced nerve damage,  glaucoma and other circulatory conditions of the
eye. This work is independent of our collaboration with Wyeth-Ayerst.


     CRF R1 Peripheral Uses

         Recent reports have suggested that  corticotropin-releasing  factor, or
CRF, plays a role in the control or modulation of the  gastrointestinal  system.
Studies have demonstrated  that central  administration of CRF acts in the brain
to inhibit emptying of the stomach while stimulating bowel activity, and suggest
that  overproduction  of  CRF  in  the  brain  may  be  a  main  contributor  to
stress-related gastrointestinal disorders.



         Irritable  bowel syndrome is a  gastrointestinal  inflammatory  disease
that  affects up to 15% of  American  adults,  mostly  women,  according  to the
International  Foundation  for  Gastrointestinal   Disorders.   Irritable  bowel
syndrome can be a lifelong, intermittent disease, involving chronic or recurrent
abdominal pain and frequent  diarrhea or  constipation,  or both.  Some patients
with  irritable  bowel  syndrome  report the onset of  symptoms  of the  disease
following a major life stress event, such as death in the family, which suggests
that the causes of  irritable  bowel  syndrome  may be  related  to  stress.  In
addition,  most irritable bowel syndrome  sufferers also experience  anxiety and
depression.  Since  researchers  believe  that CRF is the  primary  mediator  of
stress,  they have suggested that CRF R1 antagonists may provide a treatment for
irritable bowel  syndrome.  We are evaluating our proprietary CRF R1 antagonists
for treatment of irritable  bowel syndrome.  We face the risks that  preclinical
studies may not warrant initiating  clinical testing of these candidates or that
any  initial  clinical  data may not  support  continuation  of the  program and
additional clinical trials.


     CRF R2 Antagonists

         Our scientists were the first to isolate a second CRF receptor,  called
CRF R2. We believe the  distribution of CRF R2 in the brain suggests that CRF R2
could  play a role in some  forms of  anxiety  and some  eating  disorders.  Our
researchers have demonstrated that administration of a CRF R2 antagonist reduces
measures of anxiety in studies of obsessive  compulsive disorder and conditioned
fear. We are also  evaluating our proprietary CRF R2 antagonist for treatment of
a variety of eating  disorders.  We have screened our small molecule library and
conducted exploratory chemistry to identify a new series of compounds to undergo
further  study.  We face the  risks  that our work in this area will not lead to
clinical  candidates or that clinical  trials will not find our candidates to be
safe and effective.


      CRF R2 Agonists/Urocortin Agonist

         CRF R2 agonists may represent a therapeutic strategy to elevate CRF and
a related neuropeptide called urocortin.  Preliminary data indicate that CRF and
urocortin may act as central regulators of both appetite and metabolism. We have
evaluated CRF R2 agonists in various models of obesity and have observed reduced
food  intake and weight  loss.  In 1996,  we  initiated  a  three-year  research
collaboration with Eli Lilly to screen and optimize CRF R2 agonists.  In October
1999, the funded research  portion of the program was completed as scheduled and
Eli Lilly retained control of the program and exclusive rights to the compounds.
We face the risks that Eli Lilly may not initiate  further research and that, if
they  do,  the  research  may not  identify  suitable  candidate  compounds  for
development in a timely manner, or at all.


      Melanocortin Receptor Agonists/Antagonists

         Melanocortin receptors are proteins on the surface of cells, which help
regulate some body functions such as eating and skin color. To date, researchers
have  identified  a family of five  melanocortin  receptor  subtypes.  Recently,
researchers  discovered that one of the melanocortin receptor subtypes,  subtype
4, plays an important role in the mechanism regulating appetite, body weight and
insulin secretion. The subtype 4 receptor is activated by melanocyte stimulating
hormone. When melanocyte stimulating hormone is injected, it reduces food intake
and  body  weight.  Responses  have  included  the  apparent  increase  in basal
metabolic rate and lowering of serum insulin  levels.  We believe that an orally
active  subtype 4 agonist may produce the same effects and,  thus, may provide a
novel  approach to the treatment of obesity or diabetes.  We hope to identify an
orally active subtype 4 agonist compound.  However,  we may fail to do so and we
face  the  risk  that  our  melanocortin  research  will  not  lead  to  product
candidates.


      Melanin Concentrating Hormone Antagonists

         Recent studies suggest that melanin  concentrating hormone plays a role
in regulating eating behavior. Based on these findings, we believe that blocking
the effect of melanin concentrating hormone with a small molecule antagonist may
represent a novel approach to the treatment of obesity. Thus, we have identified
the melanin  concentrating hormone receptor as a compelling drug target that may



be  complementary to other  obesity/anorexia  drug targets in our drug discovery
pipeline.  We face the risk  that our  research  in this  area  will not lead to
product candidates.


     Hypocretin

         Hypocretins are peptides that  researchers  have linked to a variety of
activities, including the control of eating, cardiovascular regulation and water
intake. Recent publications have also reported that hypocretins appear to have a
critical role as regulators of sleep. Some studies point to a lack of hypocretin
as being  instrumental in the development of narcolepsy and suggest that a small
molecule  agonist  may be able to  offset  the lack of  hypocretin  and  provide
therapy  for  narcolepsy.  It  is  possible  that  the  hypocretin  system  also
contributes  to the  regulation of other  sleeping  disorders  such as insomnia,
particularly  since  administration  of excess  hypocretin into animals promotes
wakefulness.  We have screened our small molecule  library to identify  agonists
and  antagonists  for  the  hypocretin  receptors  and  are  in the  process  of
optimizing  the compounds  that resulted  from these  screens.  We will be using
these compounds to further  characterize the hypocretin system. We face the risk
that our research in this area will not lead to product candidates.


Our Discovery Technology

         We utilize a proprietary approach to small molecule drug design that we
refer to as multi-channel technology.

         Our  Multi-Channel   Discovery.   The  advent  of  molecular   biology,
culminating  recently in the cloning of the human  genome,  has opened up a vast
array of receptors as potential  targets for drug  discovery.  Over the past ten
years  numerous  new  technologies  have  been  developed  to try to  speed  the
identification of small molecule drugs. These technologies have mainly relied on
the creation of large  chemical  libraries  utilizing  combinatorial  chemistry,
automated synthesis of compounds and ultra-high throughput screening machines to
test hundreds of thousands of compounds against a particular receptor.  While we
utilize all of these  technologies,  we have taken the science to the next step,
one we call Multi-Channel Discovery or MCD.

         MCD  is  driven  by  the  power  of  traditional   medicinal  chemistry
accelerated by a suite of computational  methodologies  that guide the synthesis
of highly active small  molecules.  At the core of MCD is our  development  of a
virtual  library  (NBI-VL)   containing  over  108  molecules.   Utilizing  this
`universe'  of compounds  our chemists  sequentially  apply  computer  generated
molecular  screens to filter  compounds that will bind to the desired  receptor.
The unique  feature of MCD is that  chemists  are no longer  constrained  by the
physical  properties  of a particular  compound but rather are free to work with
thousands  of shapes  and  charges  to design  compounds  that will fit onto the
receptor target.

         The  power  of MCD  however,  lies not in its  application  to a single
receptor  as  a  drug  target  but  in  the  database  of  compounds   that  are
characterized and isolated for the next target from the same class of receptors.
Our current focus is on the most attractive receptor class in the pharmaceutical
industry, G-protein-coupled receptors (GPCR's). MCD is not a static process, but
one that becomes more powerful,  more  predictive,  and more efficient with each
subsequent use. It is an artificial intelligence system applied to drug design.

         In  connection  with our  multi-channel  technology,  we utilize  other
advanced  technologies  to  enhance  our  drug  discovery  capabilities  and  to
accelerate the drug development process. These technologies include:

         High-Throughput  Screening.  We have  assembled  a chemical  library of
diverse, low molecular weight organic molecules for lead compound identification
and we have implemented robotics screening capabilities linked to our library of
compounds that  facilitate the rapid  identification  of new drug candidates for
multiple drug targets.  In addition,  we have designed a 10,000 compound library
focused on some of our molecular  targets.  We believe that our  utilization  of
high-throughput  screening and medicinal and peptide chemistry will enable us to
rapidly identify and optimize lead molecules.



         Molecular  Biology.  Our scientists utilize novel techniques to examine
gene  expression  in  a  variety  of  cellular  systems.  We  have  developed  a
sophisticated  technique  to evaluate  the type and quantity of genes in various
cellular  systems prior to the isolation of genes. We have also developed unique
expression  vectors and cell lines that allow for the highly  efficient  protein
expression of specific genes.

         Our drug  discovery  program  creates a  significant  amount of genetic
sequence  information.  We have  developed  a  bioinformatics  system,  which we
believe will allow us to identify novel genes involved in neurodegeneration.  We
collect data with automated  instruments and we store and analyze the data using
customized computational tools.

         Gene Sequencing.  We apply integrated automated DNA sequencing and gene
identification  technology in our drug discovery program, enabling us to perform
extended  gene  analysis in a rapid,  high-throughput  format  with  independent
linkage  into  a  sequence  identification  database.  We  have  optimized  gene
sequencing  instrumentation  for  differential  display,  a  technique  that may
facilitate the rapid identification of novel genes.


Our Strategic Alliances

         One of our business  strategies  is to utilize  strategic  alliances to
enhance our development  and  commercialization  capabilities.  To date, we have
formed the following alliances:

         Janssen Pharmaceutica,  N.V. In January 1995, we entered into the first
of two research and development agreements with Janssen Pharmaceutica,  N.V., an
indirect  wholly-owned  subsidiary of Johnson & Johnson,  to  collaborate in the
discovery,   development  and   commercialization   of  small  molecule  CRF  R1
antagonists for the treatment of anxiety,  depression and substance abuse. These
collaborations  utilize  our  expertise  in cloning  and  characterizing  CRF R1
subtypes,  CRF  pharmacology  and  medicinal  chemistry.  Under the January 1995
agreement,  Janssen  funded three years of research at  Neurocrine  and received
exclusive licenses to all CRF R1 antagonist  compounds developed during the term
of the funded research or during the year  thereafter.  The term of the licenses
are for the term of the patents  licensed under the agreement.  Pursuant to this
agreement,  we have received $2.0 million in up-front  license  payments and are
eligible  to  receive  royalties  on product  sales for the term of the  patents
covering the compounds  subject to reduction for payments to third  parties.  We
may also receive royalties for products not covered by issued patents and agreed
minimum annual  royalties.  In addition,  we have the option of co-promoting the
first marketed  product from the  collaboration in North America for five years.
The 1995 agreement  will  terminate upon the expiration of the patents  covering
the  collaboration  products  but may also be  terminated  for failure by either
party  to meet  its  obligations,  bankruptcy,  or in some  circumstances,  upon
assignment  of the  agreement  by  Janssen  or us. In 1996  Janssen  selected  a
clinical  candidate from the compounds  discovered in connection  with the first
Janssen  agreement and commenced  clinical trials in Europe.  The  collaborative
research  portion of the first  Janssen  agreement was completed as scheduled in
1997.  In September  1999,  together  with  Janssen,  we entered into an amended
agreement  to expand  the  collaborative  research  and  development  efforts to
identify  additional  small  molecule  CRF  antagonists  for  the  treatment  of
psychiatric  disorders.  In connection  with the amended Janssen  agreement,  we
received an initial  payment and will receive two years of  additional  research
funding  for  our  scientists  working  in  collaboration   with  Janssen.   All
collaboration  products  identified  under the 1999 agreement are subject to the
same terms and conditions as the products arising under the 1995 agreement.

         Under the Janssen  agreements,  we are  entitled to receive up to $39.2
million for sponsored  research,  milestones and license fees,  plus  additional
amounts for potential  royalties and  reimbursement of outside costs. The amount
we are entitled to receive  includes  $14.7 million for  sponsored  research and
$2.0 million in license fees, plus up to $22.5 million in milestone payments for
the  indications of anxiety,  depression and substance  abuse, in each case upon
achievement of development  and  regulatory  goals.  As of December 31, 2000, we
have  received a total of $20.6  million,  including  $14.3 million in sponsored
research, $3.5 million in milestones,  $2.0 million in license fees and $755,000
for  reimbursement  of outside  costs.  Janssen is  responsible  for funding all
clinical development and marketing activities, including reimbursement to us for
our promotional efforts, if any. In connection with the 1995 agreement,  Johnson
& Johnson Development Corporation purchased $5.0 million of our common stock.

         Taisho  Pharmaceutical  Co., Ltd. In December  1999, we entered into an
agreement with Taisho  Pharmaceutical Co., Ltd. providing to Taisho an exclusive



option  to  obtain   European,   Asian  and  North  American   development   and
commercialization  rights for our altered peptide ligand product,  NBI-6024, for
Type 1 Diabetes.  In exchange for this option, we a received $2.0 million option
fee.  In June 2000,  Taisho  exercised  its option as to Europe and Asia,  and a
steering  committee,  comprised of delegates from both companies,  was formed to
oversee the development of NBI-6024. Pursuant to that agreement, we will receive
license fees, milestone payments and reimbursement of development  expenses.  In
addition, if the compound is ultimately commercialized, we will receive payments
on  product  sales in  Europe  and Japan  for the term of the  patents  covering
NBI-6024 subject to adjustment for payments to third parties.  In November 2000,
we expanded our collaboration with Taisho,  granting Taisho the exclusive rights
to develop  and  commercialize  NBI-6024 in North  America  and other  countries
outside of Europe and Asia. The worldwide  collaboration is valued at up to $100
million,  including all potential licensing fees, purchase fees,  milestones and
development  expenses. As of December 31, 2000, we have received a total of $4.0
million in license  fees,  $4.0 million in milestone  payments,  and $829,000 in
reimbursements  of  development  costs.  The license fees were  deferred and are
being recognized as revenues over the life of the agreement at $319,000 in 2000,
$818,000 in each of the years 2001 through 2004 and $409,000 in 2005.

         Wyeth-Ayerst  Laboratories.  Effective  January 1999, we entered into a
Collaboration and License Agreement with Wyeth-Ayerst  Laboratories,  a division
of American Home Products Corporation, relating to the research, development and
commercialization  of compounds that control  excitatory amino acid transporters
for the treatment of neurodegenerative and psychiatric diseases. Pursuant to the
agreement, we are entitled to receive up to $80.3 million for sponsored research
and milestones,  plus additional amounts for potential royalties.  The amount we
are entitled to receive consists of $11.0 million for sponsored  research over a
three-year  period,  plus  up  to  $69.3  million  in  milestone  payments  upon
achievement of certain  research,  development  and regulatory  events.  We have
granted Wyeth-Ayerst  exclusive and non-exclusive rights to our excitatory amino
acid  transporters  technology  as  well as  exclusive  rights  to any  products
developed  during  the  collaboration.  These  licenses  are for the term of the
patents  licensed.  We will receive  royalties on product sales for the terms of
the patents  covering the  collaboration  products,  subject to adjustments  for
royalties payable to other parties.  We will also receive royalties for products
that  are not the  subject  of  issued  patents.  We also  have  the  option  to
co-promote  collaboration  products in Canada and the United  States  under some
conditions.  Wyeth-Ayerst  may  terminate  the agreement if they decide that the
research  is not  successful,  if they  decide to stop the  program or if we are
acquired by another  company.  The  agreement  may also be  terminated by either
party for the other party's failure to meet its obligations  under the agreement
or  bankruptcy.  As of December  31,  2000,  we have  received  $6.0  million in
sponsored research payments, $3.0 million for the achievement of four milestones
and $50,000 in license fees,  which are being deferred and  recognized  over the
life of the agreement.

         Eli Lilly and Company.  In October 1996, we entered into a research and
license  agreement  with Eli Lilly and Company to  collaborate in the discovery,
development and  commercialization  of CRF binding protein ligand inhibitors for
the  treatment  of  central  nervous  system  disorders  including  obesity  and
dementias,  such as Alzheimer's disease, and CRF R2 agonists for central nervous
system diseases and disorders.  Under the agreement, we were entitled to receive
three years of funded research payments.  In October 1999, the funded portion of
the research program concluded as scheduled and, to our knowledge,  Eli Lilly is
not planning any specific future development  efforts.  Through October 1999, we
received a total of $17.2 million in research and development payments under the
agreement.  We  have  granted  Eli  Lilly  an  exclusive  worldwide  license  to
manufacture and market CRF binding protein ligand antagonists and CRF R2 agonist
products.  The  licenses  granted  under the  agreement  are for the term of the
patents  licensed and we are entitled to receive a royalty on product  sales for
the term of those  patents.  We have the option to  co-promote  products for the
treatment  of  dementia  in the  United  States  and to receive a portion of the
profits resulting from sales,  subject to our obligation to pay a portion of the
development  costs for such product rather than royalties.  The agreement may be
terminated  for failure by either party to meet its  obligations  or if blocking
patents prevent the development of products.

         Risks Related to Our Strategic Alliances.  We face the risks that we or
any of the  above  collaborators  may not be  successful  in  research  and drug
discovery,  that any preclinical  and clinical drug candidates  arising from the
collaborations  may not generate  commercial product candidates that have viable
clinical,  regulatory and intellectual  property profiles or that any commercial
products  arising  from  any  of  these  collaborations  may  not  enjoy  market
acceptance.  Therefore,  we may never receive  additional  milestone payments or
royalty income under any of our collaboration agreements.





Intellectual Property

         We seek to protect our lead compounds,  compound  libraries,  expressed
proteins,  synthetic organic processes,  formulations,  assays,  cloned targets,
screening technology and other technologies by filing, or by causing to be filed
on our behalf, patent applications in the United States and abroad. We have five
issued U.S.  patents,  approximately  60 pending U.S.  patent  applications  and
another  approximately  105 issued and pending foreign patents and applications.
We have  licensed,  from  institutions  such  as The  Salk  Institute,  Stanford
University,  Oregon Health Sciences University,  DOV Pharmaceuticals and others,
the rights to an  additional  30 issued U.S.  patents,  20 pending  U.S.  patent
applications,  and 50 issued and pending  foreign  filings.  Two of our European
patents are subject to opposition  proceedings.  These proceedings relate to the
CRF R2 patent and our broad patent covering immune therapeutics in diabetes.  If
successful, these opposition proceedings could reduce the breadth of some of our
proprietary  rights,  but we  believe  they  would  not  materially  impede  our
commercialization  strategy.  We face  the risk  that  one or more of the  above
patent applications may be denied. We also face the risk that our issued patents
may be challenged or  circumvented  or may otherwise not provide  protection for
any commercially viable products we develop.

         The technologies we use in our research, as well as the drug targets we
select,  may  unintentionally  infringe  the patents or violate the  proprietary
rights of third parties.  If this occurs,  we may be required to obtain licenses
to  patents  or  proprietary  rights  of others  in order to  continue  with the
commercialization  of our  products.  We are aware of pending and issued  patent
claims to melanin  concentrating  hormone  ligand and receptor,  the  hypocretin
ligand and receptor and certain uses of melanocortin subtype 4 agonists.  We are
conducting  research  in all of those areas and may  ultimately  need to license
those patents in order to proceed. In addition, we are aware of two U.S. patents
relating to IL-4  proteins  that are  controlled  by other  entities  which,  if
construed  very  broadly,  and if valid as so  construed,  could prevent us from
commercializing  our IL-4 fusion toxin  products in the United  States unless we
obtain a license,  which may not be available to us on  commercially  reasonable
terms, or at all. We do not believe that our research and development activities
as currently conducted infringe any valid issued patents.

         NBI-34060,  our leading gamma  amino-butyric  acid agonist compound for
the  treatment of insomnia,  is covered  generically  in an issued U.S.  patent,
which we licensed from DOV  Pharmaceuticals.  It is not currently covered by any
foreign  patents  of which we are aware.  The term of the U.S.  patent is due to
expire in June 2003. We intend to seek additional protection of this compound in
three ways.  First, we have filed eight U.S. and foreign patent  applications on
NBI-34060,  which could extend  patent  protection to the year 2020. We face the
risk that  these  patents  may not  issue,  or may  subsequently  be  challenged
successfully.  Second,  patent term extension under the Hatch/Waxman Patent Term
Extension Act may add patent life in the U.S.  beyond the June 2003  expiration,
depending  on the length of clinical  trials and other  factors  involved in the
filing of a new drug  application.  Third, in addition to this potential  patent
protection,  the United  States,  the European  Union and Japan all provide data
protection  for new medicinal  compounds.  If this  protection is available,  no
competitor  may use the  original  applicant's  data as the  basis of a  generic
marketing  application  during the  period of data  protection.  This  period of
exclusivity is five years in the United States, six years in Japan and six to 10
years in the European  Union,  measured  from the date of FDA, or  corresponding
foreign, approval.


In-Licensed Technology

         We have  in-licensed  the  following  technologies  to  complement  our
ongoing  clinical and research  programs.  Most of these licenses extend for the
term of the related patent and contain customary royalty,  termination and other
provisions.

o        In October  2000,  we licensed  nonexclusive  rights to GT1-1 cell line
         from The Salk Institute.

o        In August  2000,  we licensed  nonexclusive  rights to CRF R1 deficient
         mice from the Research Development Foundation.

o        In July 2000, we licensed  exclusive  rights to melanocortin  subtype 3
         receptor knock-out mice from Oregon Health Sciences University.




o        In  August  1999,  we  licensed   nonexclusive   rights  to  the  human
         gonadotropin-releasing  hormone  receptor  from Mount  Sinai  School of
         Medicine.

o        In January  1999,  we licensed  exclusive  worldwide  rights to patents
         relating to excitatory  amino acid  transporters  and  melanocortin 1-5
         from Oregon Health Sciences University.

o        In  December  1998,  we  licensed  nonexclusive  rights  to  technology
         covering  melanocortin  subtype  4  receptor  from  the  University  of
         Michigan.

o        In June 1998, we licensed  exclusive  worldwide  rights to our sedative
         compound, NBI-34060, from DOV Pharmaceuticals, Inc.

o        In May 1998, we licensed exclusive worldwide rights to patents covering
         NBI-3001, our IL-4 fusion toxin, from the National Institutes of Health
         and inventors Ira Pastan and David Fitzgerald.

o        In November 1996, we licensed exclusive  worldwide rights to technology
         directed  to  peptide  therapeutics  for the  treatment  of  autoimmune
         disease from Trustees of Dartmouth College.

o        In October 1997, we licensed co-exclusive rights to technology relating
         to the prevention of diabetes from University Technology Corporation.

o        In November 1994, we licensed exclusive  worldwide rights to technology
         relating to treatment of multiple  sclerosis  using peptide  analogs of
         myelin basic protein from Stanford University.

o        In November 1993, we licensed exclusive worldwide rights to CRF R1 from
         the Salk Institute for Biological Studies.


Manufacturing

         We currently  rely, and will continue to rely for at least the next few
years, on contract manufacturers to produce sufficient quantities of our product
candidates  for  use  in  our  preclinical  and  anticipated   clinical  trials.
Manufacturers  of our  NBI-34060 and CRF  antagonist  compounds  clinical  trial
material  include  Albany   Molecular   Research,   Cedarburg   Pharmaceuticals,
Organichem  Corporation and Pharmaceutics  International.  Cook Pharmaceuticals,
Covance Biotechnology Services, Polypeptide Laboratories,  Primedica and Pyramid
Laboratories manufacture our altered peptide ligands,  NBI-6024 and NBI-5788 and
our recombinant protein NBI-3001.  We also rely, and intend to continue to rely,
on third parties to provide the components of these product candidates,  such as
proteins, peptides, phospholipids, small molecules and bulk chemical materials.

         There is  currently  a  limited  supply  of some of  these  components.
Furthermore,  the contract  manufacturers  that we have  identified to date only
have limited experience at manufacturing,  formulating,  analyzing and packaging
our product candidates in quantities  sufficient for conducting  clinical trials
or for commercialization.

         If we are unable to  establish  and maintain  relationships  with third
parties for manufacturing  sufficient  quantities of our product  candidates and
their components that meet our planned time and cost parameters,  it could delay
the development and timing of our clinical trials.


Marketing and Sales

     We currently  have no sales,  marketing or  distribution  capabilities.  In
order to commercialize any of our product candidates,  we must either internally
develop sales, marketing and distribution capabilities or make arrangements with
third  parties  to  perform  these  services.  We  intend  to sell,  market  and
distribute some products directly and intend to rely on relationships with third
parties to sell,  market and distribute other products.  Under our collaboration
agreements with Janssen, Wyeth-Ayerst and Eli Lilly, we may have the opportunity



to co-promote our products in the United  States.  To market any of our products
directly,  we must develop a marketing and sales force with technical  expertise
and with supporting distribution capabilities, none of which we currently have.


Government Regulation

         Regulation by government  authorities  in the United States and foreign
countries is a significant factor in the development,  manufacture and marketing
of our  proposed  products and in our ongoing  research and product  development
activities.  All of our products will require regulatory  approval by government
agencies prior to commercialization.  In particular,  human therapeutic products
are  subject to  rigorous  preclinical  studies  and  clinical  trials and other
approval  procedures of the FDA and similar  regulatory  authorities  in foreign
countries.  Various  federal and state statutes and  regulations  also govern or
influence testing,  manufacturing,  safety, labeling, storage and record-keeping
related to such  products and their  marketing.  The process of obtaining  these
approvals and the subsequent substantial compliance with appropriate federal and
state statutes and regulations  require the expenditure of substantial  time and
financial resources.

         Preclinical  studies are generally  conducted in laboratory  animals to
evaluate the  potential  safety and the efficacy of a product.  Drug  developers
submit  the  results  of  preclinical  studies  to  the  FDA  as a  part  of  an
investigative new drug  application,  which must be approved before we can begin
clinical  trials in  humans.  Typically,  clinical  evaluation  involves  a time
consuming and costly three-phase process.


     Phase I     Clinical  trials are  conducted  with a small number of
                 subjects  to  determine  the  early  safety  profile,   maximum
                 tolerated  dose and  pharmacokinetics  of the  product in human
                 volunteers.

     Phase II    Clinical  trials are conducted  with groups of patients
                 afflicted  with  a  specific  disease  in  order  to  determine
                 preliminary efficacy,  optimal dosages and expanded evidence of
                 safety.

     Phase III   Large-scale,   multi-center,   comparative  trials  are
                 conducted  with  patients  afflicted  with a target  disease in
                 order to provide  enough data to demonstrate  with  substantial
                 evidence the efficacy and safety required by the FDA.

         The FDA closely  monitors  the  progress of each of the three phases of
clinical  trials  that are  conducted  in the  United  States  and  may,  at its
discretion,  reevaluate,  alter, suspend or terminate the testing based upon the
data  accumulated  to that point and the FDA's  assessment  of the  risk/benefit
ratio to the patient.  To date we have conducted many of our clinical  trials in
Europe and South Africa.

         Once Phase III trials are completed, drug developers submit the results
of preclinical  studies and clinical trials to the FDA in the form of a new drug
application,  or a  biologics  licensing  application  for  approval to commence
commercial  sales. In response,  the FDA may grant marketing  approval,  request
additional  information or deny the  application if the FDA determines  that the
application does not meet regulatory approval criteria. FDA approvals may not be
granted on a timely basis,  or at all.  Furthermore,  the FDA may prevent a drug
developer  from  marketing a product under a label for its desired  indications,
which may impair commercialization of the product. Similar regulatory procedures
must also be complied with in countries outside the United States.

         If the  FDA  approves  the  new  drug  application,  the  drug  becomes
available for physicians to prescribe in the United States. After approval,  the
drug developer must submit periodic reports to the FDA,  including  descriptions
of any adverse reactions reported. The FDA may request additional studies, known
as Phase IV, to evaluate long-term effects.

         In  addition to studies  requested  by the FDA after  approval,  a drug
developer  may conduct  other  trials and studies to explore use of the approved
compound  for  treatment  of new  indications.  The purpose of these  trials and
studies and related  publications  is to broaden the  application and use of the
drug and its acceptance in the medical community.



     Orphan Drug Designation

         The FDA may grant orphan drug  designation to drugs intended to treat a
"rare  disease or  condition,"  which is generally a disease or  condition  that
affects  fewer than  200,000  individuals  in the  United  States.  Orphan  drug
designation must be requested before  submitting a new drug  application.  After
the FDA grants orphan drug designation,  the generic identity of the therapeutic
agent and its  potential  orphan use are disclosed  publicly by the FDA.  Orphan
drug  designation  does not convey any advantage in, or shorten the duration of,
the regulatory  review and approval  process.  If a product that has orphan drug
designation  subsequently  receives FDA approval for the indication for which it
has orphan drug  designation,  the  product is  entitled to orphan  exclusivity,
which means the FDA may not approve  any other  applications  to market the same
drug for the same indication,  except in very limited  circumstances,  for seven
years.  Our IL-4  fusion  toxin  product  candidate  has  received  orphan  drug
designation from the FDA for astrocytic glioma.


     Approvals Outside the United States

         We will have to  complete an  approval  process  similar to that in the
United States in virtually every foreign target market for our products in order
to  commercialize  our  product  candidates  in those  countries.  The  approval
procedure  and the time  required for approval  vary from country to country and
may involve additional testing. Foreign approvals may not be granted on a timely
basis, or at all. In addition, regulatory approval of prices is required in most
countries  other than the  United  States.  We face the risk that the  resulting
prices  would be  insufficient  to generate an  acceptable  return to us, or our
corporate collaborators.


        Fast Track Designation

         Congress enacted the Food and Drug Administration  Modernization Act of
1997, in part, to ensure the timely  availability  of safe and effective  drugs,
biologics  and  medical  devices by  expediting  the FDA review  process for new
products.  The Food and Drug  Administration  Modernization  Act  establishes  a
statutory program for the approval of so-called fast track products. The new law
defines  a fast  track  product  as a new  drug  or  biologic  intended  for the
treatment  of a serious or  life-threatening  condition  that  demonstrates  the
potential to address  unmet  medical  needs for such a condition.  Under the new
fast track program, the sponsor of a new drug or biologic may request the FDA to
designate  the drug or  biologic  as a fast  track  product  at any time  during
clinical  development  of  the  product.   Fast-track  designation  provides  an
expedited review of a product, which accelerates FDA review.

         We may seek  fast  track  designation  to  secure  expedited  review of
appropriate  product  candidates.  We can never be sure that we will obtain fast
track  designation.  We cannot predict the ultimate  impact,  if any, of the new
fast track  process on the timing or  likelihood  of FDA  approval of any of our
potential  products.  We  received  fast track  designation  for our IL-4 fusion
toxin.


Competition

         The  biotechnology and  pharmaceutical  industries are subject to rapid
and  intense   technological  change.  We  face,  and  will  continue  to  face,
competition  in the  development  and marketing of our product  candidates  from
biotechnology and pharmaceutical  companies,  research institutions,  government
agencies and academic institutions. Competition may also arise from, among other
things:

o        other drug development technologies;

o        methods of preventing  or reducing the incidence of disease,  including
         vaccines; and

o        new small molecule or other classes of therapeutic agents.




         Developments   by  others  may  render  our   product   candidates   or
technologies  obsolete  or  noncompetitive.  We are  performing  research  on or
developing  products for the treatment of several disorders  including  anxiety,
depression,  insomnia,  malignant  glioma,  other  forms of cancer and  multiple
sclerosis.

         In the area of anxiety  disorders,  our product candidates will compete
with well-established products such as Valium(R),  marketed by Hoffman-La Roche,
Xanax(R),    marketed   by   Pharmacia    Upjohn,    Buspar(R),    marketed   by
Bristol-Myers-Squibb, and others.

         We are also developing products for depression, which will compete with
well-established  products in the  antidepressant  class,  including  Prozac(R),
marketed by Eli Lilly,  Zoloft(R) marketed by Pfizer, and Paxil(R),  marketed by
GlaxoSmith Kline. Certain technologies under development by other pharmaceutical
companies  could result in additional  commercial  treatments for depression and
anxiety.  In addition,  a number of companies  are also  conducting  research on
molecules to block CRF,  which is the same  mechanism of action  employed by our
compounds.

         We are also  developing a gamma  amino-butyric  acid receptor  agonist,
NBI-34060,  for the  treatment of insomnia.  Ambien(R) and Sonata(R) are already
marketed for the  treatment of insomnia by  Pharmacia/Sanofi  and American  Home
Products, respectively.

         Guilford  Pharmaceuticals'  Gliadel(R),  approved  for use in a type of
brain  cancers known as recurrent  glioblastoma  multiforme,  would  potentially
compete with our IL-4 fusion toxin product, NBI-3001, if our product is approved
by the FDA.  Temozolomide,  marketed by Schering  Plough,  is approved in Europe
only for recurrent glioblastoma  multiforme,  where it may also compete with our
IL-4 fusion toxin product.

         We are also  pursuing  development  of NBI-3001  for the  treatment  of
peripheral solid tumors,  such as kidney cancer and non-small-cell  lung cancer.
Proleukin(R) is marketed by Chiron for the treatment of kidney cancer,  and drug
treatments  for  non-small-cell  lung cancer include  Platinol(R)  and Taxol(R),
which are marketed by Bristol-Myers-Squibb,  and Gemzar(R), which is marketed by
Eli Lilly.

         Products that may compete with NBI-5788, our altered peptide ligand for
multiple  sclerosis,  include  Betaseron(R)  and  Avonex(R),  similar  forms  of
beta-interferon  marketed  by  Berlex  BioSciences  and  Biogen,   respectively.
Copaxone(R),  a peptide polymer marketed by Teva, has also been approved for the
marketing in the United States and certain other  countries for the treatment of
multiple sclerosis.

         There are a number of competitors to products in our research pipeline.
Lupron Depot(R), marketed by Takeda-Abbott Pharmaceuticals, Zoladex(R), marketed
by   AstraZeneca,   and   Synarel(R),   marketed  by   Pharmacia   Upjohn,   are
gonadotropin-releasing  hormone peptide agonists that have been approved for the
treatment of prostate  cancer,  endometriosis,  infertility,  breast  cancer and
central  precocious  puberty.  In  addition,   peptide  antagonists,   including
Abarelix(R)  and  Antagon  Injection(R),  are  under  development  by Amgen  and
Organon,  respectively,  for these indications. These drugs may compete with any
small molecule  gonadotropin-releasing  hormone antagonists we develop for these
indications.

         Anti-obesity  therapeutics  currently available include Xenical(R) from
Roche Laboratories and Meridia(R) from Knoll Pharmaceuticals.  If one or more of
these products or programs are successful, it may reduce or eliminate the market
for our products.

         Compared to us, many of our competitors and potential  competitors have
substantially greater:

o        capital resources;

o        research and development resources, including personnel and technology;

o        regulatory experience;

o        preclinical study and clinical testing experience;

o        manufacturing and marketing experience; and



o        production facilities.

         Any of these  competitive  factors could harm our business,  prospects,
financial condition and results of operations, which could negatively impact our
stock price.


Insurance

         The Company maintains product liability  insurance for clinical trials.
The  Company  intends to expand its  insurance  coverage  to include the sale of
commercial   products  if  marketing   approval  is  obtained  for  products  in
development. However, insurance coverage is becoming increasingly expensive, and
no assurance  can be given that the Company  will be able to maintain  insurance
coverage at a reasonable  cost or in  sufficient  amounts to protect the Company
against losses due to liability. There can also be no assurance that the Company
will be able to obtain  commercially  reasonable product liability insurance for
any products  approved for marketing.  A successful  product  liability claim or
series of claims  brought  against  the  Company  could have a material  adverse
effect on its business, financial condition and results of operations.


Employees

         As of  December  31,  2000,  we had 188  employees,  consisting  of 179
full-time and 9 part-time employees.  Of the full-time employees,  approximately
63 hold Ph.D., M.D. or equivalent degrees. None of our employees are represented
by a collective bargaining arrangement, and we believe our relationship with our
employees  is good.  We are highly  dependent  on the  principal  members of our
management and scientific staff. If we were to lose the services of any of these
personnel,  we  might  not  be  able  to  achieve  our  development  objectives.
Furthermore,  recruiting and retaining qualified scientific personnel to perform
research  and  development  work in the  future  will  also be  critical  to our
success.  We face  the  risk  that we may not be  able  to  attract  and  retain
personnel  on  acceptable  terms  given  the  competition  among  biotechnology,
pharmaceutical and health care companies,  universities and non-profit  research
institutions for experienced scientists.  In addition, we rely on members of our
Scientific  Advisory Board and a significant  number of consultants to assist us
in formulating our research and development strategies.


                                  RISK FACTORS


Risks Related to the Company

         We have a history of losses and expect to incur substantial  losses and
negative  operating  cash  flows for the  foreseeable  future,  and we may never
achieve  or  maintain  profitability.  Since  our  inception,  we have  incurred
significant net losses, including net losses of $28.8 million in the period from
January 1, 2000  through  December 31,  2000.  As a result of ongoing  operating
losses, we had an accumulated  deficit of $70.5 million as of December 31, 2000.
We  are  not  currently  profitable.  Even  if  we  succeed  in  developing  and
commercializing  one or more of our drugs, we expect to incur substantial losses
for the foreseeable  future and may never become  profitable.  We also expect to
continue to incur significant  operating and capital expenditures and anticipate
that our expenses will increase substantially in the foreseeable future as we:

o        seek regulatory approvals for our product candidates;

o        develop, formulate, manufacture and commercialize our drugs;

o        implement additional internal systems and infrastructure; and

o        hire additional clinical and scientific personnel.



         We also expect to  experience  negative  cash flow for the  foreseeable
future as we fund our operating losses and capital expenditures. As a result, we
will  need  to  generate   significant   revenues   to  achieve   and   maintain
profitability.  We may not be able to generate these revenues,  and we may never
achieve  profitability  in the  future.  Our  failure  to  achieve  or  maintain
profitability could negatively impact the market price of our common stock.

         If we cannot  raise  additional  funding,  we may be unable to complete
development  of our product  candidates.  We may require  additional  funding to
continue our research and product development  programs,  including  preclinical
testing and clinical trials of our product  candidates,  for operating expenses,
and to pursue regulatory  approvals for product candidates.  We also may require
additional funding to establish  manufacturing and marketing capabilities in the
future. We believe that our existing capital  resources,  together with interest
income and future payments due under our strategic alliances, will be sufficient
to satisfy our current and projected funding  requirements for at least the next
12 months.  However,  these resources might be insufficient to conduct  research
and development  programs as planned. If we cannot obtain adequate funds, we may
be required to curtail significantly one or more of our research and development
programs or obtain funds through  arrangements  with corporate  collaborators or
others that may require us to relinquish  rights to some of our  technologies or
product candidates.

         Our future capital requirements will depend on many factors, including:

o        continued scientific progress in our research and development programs;

o        the magnitude of our research and development programs;

o        progress with preclinical testing and clinical trials;

o        the time and costs involved in obtaining regulatory approvals;

o        the costs  involved  in filing and  pursuing  patent  applications  and
         enforcing patent claims;

o        competing technological and market developments;

o        the establishment of additional strategic alliances;

o        the  cost  of   manufacturing   facilities  and  of   commercialization
         activities and arrangements; and

o        the cost of product in-licensing and any possible acquisitions.

         We intend to seek additional funding through strategic  alliances,  and
may seek  additional  funding through public or private sales of our securities,
including  equity  securities.  In addition,  we have leased  equipment  and may
continue to pursue  opportunities  to obtain  additional  debt  financing in the
future.  However,  additional equity or debt financing might not be available on
reasonable  terms,  if at all,  and any  additional  equity  financings  will be
dilutive to our stockholders.

         Because  the  development  of our  product  candidates  is subject to a
substantial  degree  of  technological  uncertainty,   we  may  not  succeed  in
developing any of our product  candidates.  All of our product candidates are in
research or development and we do not expect any of our product candidates to be
commercially  available  for the  foreseeable  future,  if at all.  Only a small
number of research and development  programs  ultimately  result in commercially
successful drugs. Potential products that appear to be promising at early stages
of development  may not reach the market for a number of reasons.  These reasons
include the possibilities that the potential products may:

o        be found  ineffective or cause harmful side effects during  preclinical
         studies or clinical trials;

o        fail to receive necessary regulatory approvals;



o        be precluded  from  commercialization  by  proprietary  rights of third
         parties;

o        be difficult to manufacture on a large scale; or

o        be uneconomical or fail to achieve market acceptance.

     If any of these potential problems occurs, we may never successfully market
any products.

         We may not receive  regulatory  approvals  for our product  candidates.
Regulation by government  authorities in the United States and foreign countries
is a significant  factor in the  development,  manufacture  and marketing of our
proposed   products  and  in  our  ongoing  research  and  product   development
activities.  All of our products are in research and development and we have not
yet requested or received  regulatory approval to commercialize any product from
the FDA or any other  regulatory  body. The process of obtaining these approvals
and the subsequent  substantial  compliance with  appropriate  federal and state
statutes  and  regulations  require  spending  substantial  time  and  financial
resources.  If we fail or our  collaborators  or  licensees  fail to obtain,  or
encounter  delays in obtaining or maintaining,  regulatory  approvals,  it could
adversely  affect the  marketing  of any  products  we  develop,  our ability to
receive product or royalty revenues and our liquidity and capital resources.

         In  particular,  human  therapeutic  products  are  subject to rigorous
preclinical testing and clinical trials and other approval procedures of the FDA
and similar  regulatory  authorities in foreign  countries.  Various federal and
state statutes and regulations also govern or influence testing,  manufacturing,
safety, labeling, storage and record-keeping related to these products and their
marketing.  Any delay in, or suspension  of, our clinical  trials will delay the
filing of our new drug applications with the FDA and, ultimately, our ability to
commercialize our drugs and generate product revenues.

         In connection with our clinical trials, we face the risks that:

o        we or the FDA may suspend the trials;

o        we may  discover  that a  product  candidate  may  cause  harmful  side
         effects;

o        the results may not replicate the results of earlier, smaller trials;

o        the results may not be statistically significant;

o        patient recruitment may be slower than expected; and

o        patients may drop out of the trials.

         In addition, we depend on independent clinical investigators to conduct
our clinical trials under their agreements with us. These  investigators are not
our employees and we cannot  control the amount or timing of resources that they
devote to our programs.  If independent  investigators fail to devote sufficient
time and resources to our drug development  programs, or if their performance is
substandard,  it  will  delay  the  approval  of our  FDA  applications  and our
introductions of new drugs. These investigators may also have relationships with
other  commercial  entities,  some of which may compete with us. If  independent
investigators  assist  our  competitors  at  our  expense,  it  could  harm  our
competitive position.

         Also,  late stage  clinical  trials are often  conducted  with patients
having the most  advanced  stages of  disease.  During the course of  treatment,
these patients can die or suffer other adverse  medical effects for reasons that
may not be  related  to the  pharmaceutical  agent  being  tested  but which can
nevertheless adversely affect clinical trial results.

         We depend on continuing our current strategic  alliances and developing
additional  strategic  alliances to develop and commercialize our compounds.  We
depend upon our corporate collaborators to provide adequate funding for a number
of our  programs.  Under these  arrangements,  our corporate  collaborators  are
responsible for:



o        selecting compounds for subsequent development as drug candidates;

o        conducting  preclinical  studies  and  clinical  trials  and  obtaining
         required regulatory approvals for these drug candidates; and

o        manufacturing and commercializing any resulting drugs.

         Our  strategy  for  developing  and  commercializing  our  products  is
dependent  upon  maintaining  our  current  arrangements  and  establishing  new
arrangements with research collaborators, corporate collaborators and others. We
currently have collaborations with Janssen Pharmaceutica,  Wyeth-Ayerst,  Taisho
Pharmaceutical  and  Eli  Lilly.  Because  we  rely  heavily  on  our  corporate
collaborators, our development of our projects would be substantially delayed if
our collaborators:

o        fail to select a compound we have discovered for subsequent development
         into marketable products;

o        fail to gain the requisite regulatory approvals of these products;

o        do not successfully commercialize products that we originate;

o        do not conduct their collaborative activities in a timely manner;

o        do not devote  sufficient time and resources to our partnered  programs
         or potential products;

o        terminate their alliances with us;

o        develop,  either alone or with others,  products  that may compete with
         our products;

o        dispute  our  respective  allocations  of  rights  to any  products  or
         technology developed during our collaborations; or

o        merge with a third party that may wish to terminate the collaboration.

         These   issues   and   possible   disagreements   with  our   corporate
collaborators could lead to delays in the collaborative research, development or
commercialization of many of our product candidates. Furthermore,  disagreements
with these parties could require or result in litigation or  arbitration,  which
would be  time-consuming  and  expensive.  If any of these issues arise,  it may
delay the filing of our new drug applications and, ultimately, our generation of
product revenues.

         We have no manufacturing capabilities.  If third-party manufacturers of
our product  candidates  fail to devote  sufficient  time and  resources  to our
concerns,  or if their  performance  is  substandard,  our  clinical  trials and
product introductions may be delayed and our costs may rise.

         We have in the past  utilized,  and  intend  to  continue  to  utilize,
third-party  manufacturers  to produce the drug compounds we use in our clinical
trials and for the potential  commercialization of our future products.  We have
no  experience  in  manufacturing  products for  commercial  purposes and do not
currently have any manufacturing facilities.  Consequently, we depend on several
contract  manufacturers  for all  production  of products  for  development  and
commercial  purposes.   If  we  are  unable  to  obtain  or  retain  third-party
manufacturers,   we  will  not  be  able  to  commercialize  our  products.  The
manufacture  of our  products  for clinical  trials and  commercial  purposes is
subject to specific FDA regulations.  In addition, our third-party manufacturers
may not comply with FDA regulations  relating to manufacturing  our products for
clinical trials and commercial purposes or other regulatory  requirements now or
in the future.  Our  reliance on contract  manufacturers  also exposes us to the
following risks:

o        Contract  manufacturers may encounter  difficulties in achieving volume
         production,  quality  control  and  quality  assurance,  and  also  may
         experience shortages in qualified personnel.  As a result, our contract
         manufacturers might not be able to meet our clinical schedules.



o        Switching   manufacturers  may  be  difficult  because  the  number  of
         potential  manufacturers is limited.  It may be difficult or impossible
         for us to find a replacement  manufacturer quickly on acceptable terms,
         or at all.

o        Our contract  manufacturers may not perform as agreed or may not remain
         in the  contract  manufacturing  business  for  the  time  required  to
         successfully produce, store and distribute our products.

o        Drug   manufacturers  are  subject  to  ongoing  periodic   unannounced
         inspection by the FDA, the Drug Enforcement  Agency,  and corresponding
         state  agencies to ensure  strict  compliance  with good  manufacturing
         practices and other government  regulations and  corresponding  foreign
         standards.  We do not  have  control  over  third-party  manufacturers'
         compliance with these regulations and standards.

         Our current  dependence  upon third parties for the  manufacture of our
products may harm our profit margin,  if any, on the sale of our future products
and our  ability to develop and  deliver  products  on a timely and  competitive
basis.

         If we are unable to retain and recruit  qualified  scientists or if any
of our key senior executives discontinue their employment with us, it will delay
our development efforts. We are highly dependent on the principal members of our
management  and scientific  staff.  The loss of any of these people could impede
the  achievement  of our  development  objectives.  Furthermore,  recruiting and
retaining  qualified  scientific  personnel to perform  research and development
work in the future  will also be critical  to our  success.  We may be unable to
attract and retain  personnel on acceptable  terms given the  competition  among
biotechnology,  pharmaceutical  and  health  care  companies,  universities  and
non-profit research  institutions for experienced  scientists.  In addition,  we
rely on members of our  Scientific  Advisory  Board and a significant  number of
consultants to assist us in formulating our research and  development  strategy.
All of our consultants and members of the Scientific Advisory Board are employed
by  employers  other  than us.  They may have  commitments  to, or  advisory  or
consulting  agreements with, other entities that may limit their availability to
us.

         We may be subject to claims that we, or our employees,  have wrongfully
used or  disclosed  alleged  trade  secrets  of their  former  employers.  As is
commonplace  in the  biotechnology  industry,  we  employ  individuals  who were
previously  employed  at  other   biotechnology  or  pharmaceutical   companies,
including our competitors or potential  competitors.  Although no claims against
us are currently pending, we may be subject to claims that these employees or we
have  inadvertently  or  otherwise  used or  disclosed  trade  secrets  or other
proprietary  information of their former employers.  Litigation may be necessary
to defend against these claims.  Even if we are successful in defending  against
these claims,  litigation could result in substantial costs and be a distraction
to management.

         We  have  no  marketing   experience,   sales  force  or   distribution
capabilities  and,  if our  products  are  approved,  we  may  not  be  able  to
commercialize  them  successfully.   Although  we  do  not  currently  have  any
marketable  products,  our ability to produce revenues ultimately depends on our
ability  to sell our  products  if and when  they are  approved  by the FDA.  We
currently have no experience in marketing or selling pharmaceutical products and
we do not have a marketing and sales staff or distribution  capabilities.  If we
fail to establish  successful  marketing and sales capabilities or fail to enter
into successful marketing  arrangements with third parties, our product revenues
will suffer.

         Governmental  and third-party  payers may subject our products to sales
and  pharmaceutical  pricing  controls that could limit our product revenues and
delay profitability. The continuing efforts of government and third-party payers
to contain or reduce the costs of health care through  various  means may reduce
our potential  revenues.  These payers' efforts could decrease the price that we
receive for any  products we may  develop and sell in the future.  In  addition,
third-party insurance coverage may not be available to patients for any products
we  develop.  If  government  and  third-party  payers do not  provide  adequate
coverage and  reimbursement  levels for our products,  or if price  controls are
enacted, our product revenues will suffer.

         If  physicians  and  patients  do not accept our  products,  we may not
recover our  investment.  The  commercial  success of our products,  if they are
approved  for  marketing,  will depend upon the medical  community  and patients
accepting our products as being safe and effective. The market acceptance of our
products could be affected by a number of factors, including:



o        the timing of receipt of marketing approvals;

o        the safety and efficacy of the products;

o        the emergence of equivalent or superior products; and

o        the cost-effectiveness of the products.

     If the medical community and patients do not ultimately accept our products
as being safe and effective, we may not recover our investment.


Risks Related to Our Industry

         We  face  intense   competition   and  if  we  are  unable  to  compete
effectively,  the  demand  for  our  products,  if  any,  may  be  reduced.  The
biotechnology  and  pharmaceutical  industries  are subject to rapid and intense
technological  change.  We face,  and will continue to face,  competition in the
development and marketing of our product candidates from academic  institutions,
government agencies,  research institutions and biotechnology and pharmaceutical
companies. Competition may also arise from, among other things:

o        other drug development technologies;

o        methods of preventing  or reducing the incidence of disease,  including
         vaccines; and

o        new small molecule or other classes of therapeutic agents.

         Developments   by  others  may  render  our   product   candidates   or
technologies  obsolete  or  noncompetitive.  We are  performing  research  on or
developing  products for the treatment of several disorders  including  anxiety,
depression,  insomnia,  malignant  glioma,  other  forms of cancer and  multiple
sclerosis,  and there are a number of  competitors  to products in our  research
pipeline.  If one or more of these  products or  programs  are  successful,  the
market for our products may be reduced or eliminated.

         Compared to us, many of our competitors and potential  competitors have
substantially greater:

o        capital resources;

o        research and development resources, including personnel and technology;

o        regulatory experience;

o        preclinical study and clinical testing experience;

o        manufacturing and marketing experience; and

o        production facilities.

         Any of these competitive  factors could reduce demand for our products.
For more specific  information  about the  competition  we face,  please see the
section "Business" under the subheading "Competition".

         If we are unable to protect our intellectual  property, our competitors
could develop and market  products  based on our  discoveries,  which may reduce
demand for our products.  Our success will depend on our ability to, among other
things:

o        obtain patent protection for our products;



o        preserve our trade secrets;

o        prevent third parties from infringing upon our proprietary rights; and

o        operate without infringing upon the proprietary rights of others,  both
         in the United States and internationally.

         Because of the substantial  length of time and expense  associated with
bringing new products through the development and regulatory  approval processes
in  order  to  reach  the  marketplace,   the  pharmaceutical   industry  places
considerable  importance on obtaining patent and trade secret protection for new
technologies,  products  and  processes.  Accordingly,  we intend to seek patent
protection for our proprietary  technology and compounds.  However,  we face the
risk that we may not obtain any of these  patents and that the breadth of claims
we obtain,  if any,  may not  provide  adequate  protection  of our  proprietary
technology or compounds.

         We also rely upon unpatented trade secrets and improvements, unpatented
know-how and  continuing  technological  innovation  to develop and maintain our
competitive  position,  which we seek to protect,  in part,  by  confidentiality
agreements with our commercial collaborators, employees and consultants. We also
have invention or patent assignment  agreements with our employees and some, but
not all,  of our  commercial  collaborators  and  consultants.  However,  if our
employees,  commercial collaborators or consultants breach these agreements,  we
may not have  adequate  remedies for any such breach,  and our trade secrets may
otherwise become known or independently discovered by our competitors.

         We may not be able to adequately  enforce any of our patents to protect
our proprietary technology and compounds.  Litigation may be necessary to defend
against or assert  infringement  claims to enforce  our  issued  patents  and to
protect our trade secrets or know-how, or to determine the scope and validity of
the  proprietary  rights of others.  Two of our European  patents are subject to
opposition proceedings,  which, if successful,  could reduce the breadth of some
of   our    proprietary    rights.    These    proceedings    relate    to   our
corticotropin-releasing  factor  receptor  patent and our broad patent  covering
immune therapeutics in diabetes. In addition,  interference proceedings declared
by the United States  Patent and Trademark  Office may be necessary to determine
the priority of inventions  with respect to our patent  applications or those of
our  licensors.  Litigation or  interference  proceedings  may fail and, even if
successful, may result in substantial costs and be a distraction to management.

         The  technologies we use in our research as well as the drug targets we
select may  unintentionally  infringe  the  patents or violate  the  proprietary
rights of third parties. In such cases, we may be required to obtain licenses to
patents or  proprietary  rights of others in order to continue to  commercialize
our products.  However, we may not be able to obtain any licenses required under
any patents or proprietary  rights of third parties on acceptable  terms,  or at
all. If we do not obtain those licenses,  we could  encounter  delays in product
introductions  while we attempt to design around those patents, or we could find
that we are unable to develop,  manufacture  or sell  products  requiring  those
licenses.  We are aware of pending and issued  patent  claims to certain uses of
some of the types of compounds we are developing.

         If we are unable to resolve third-party disputes regarding the validity
of our patents or our alleged  infringement of other third parties' patents,  we
may not be able to sell some or all of our products.  For more information about
our  intellectual  property,   please  see  the  section  "Business"  under  the
subheading "Intellectual Property".

         We face  potential  product  liability  exposure  far in  excess of our
limited insurance coverage. The use of any of our potential products in clinical
trials,  and the sale of any  approved  products,  may  expose  us to  liability
claims. These claims might be made directly by consumers, health care providers,
pharmaceutical  companies  or others  selling  our  products.  We have  obtained
limited  product  liability  insurance  coverage for our clinical  trials in the
amount of $5 million per occurrence  and $5 million in the  aggregate.  However,
insurance coverage is becoming increasingly expensive, and we may not be able to
maintain  insurance  coverage at a reasonable  cost or in sufficient  amounts to
protect us against  losses due to  liability.  We intend to expand our insurance
coverage  to include  the sale of  commercial  products  if we obtain  marketing
approval for product  candidates in development,  but we may be unable to obtain
commercially  reasonable  product liability  insurance for any products approved
for marketing. On occasion,  juries have awarded large judgments in class action



lawsuits  based on drugs  that had  unanticipated  side  effects.  A  successful
product  liability  claim or series of claims brought  against us would decrease
our cash reserves and could cause our stock price to fall.

         Our activities involve hazardous materials and we may be liable for any
resulting  contamination  or  injuries.  Our  research  activities  involve  the
controlled  use  of  hazardous  materials.  We  cannot  eliminate  the  risk  of
accidental  contamination or injury from these materials. If an accident occurs,
a court may hold us liable for any resulting damages,  which may reduce our cash
reserves and force us to seek additional financing.


                            SCIENTIFIC ADVISORY BOARD

         We have assembled a Scientific  Advisory Board that currently  consists
of 12 individuals.  Members of our Scientific  Advisory Board are leaders in the
fields of  neurobiology,  immunology,  endocrinology,  psychiatry  and medicinal
chemistry.  Our Scientific Advisory Board members meet at least yearly to advise
us in the selection, implementation and prioritization of our research programs.
Some  members  meet more  frequently  to advise us with  regard to our  specific
programs.

         Our  Scientific  Advisory  Board  presently  consists of the  following
individuals:

         Susan G. Amara,  Ph.D., a Senior  Scientist and Professor at the Vollum
Institute  for Advanced  Biomedical  Research,  is an expert on the cellular and
molecular  biology  of  neurotransmitter  transporters,  excitatory  amino  acid
transporter  structure and  regulation and signaling  roles of  neurotransmitter
transporters.

         Floyd   E.   Bloom,   M.D.,   is   Chairman   of  the   Department   of
Neuropharmacology   at  The  Scripps  Research   Institute.   Dr.  Bloom  is  an
internationally  recognized  expert  in  the  fields  of  neuropharmacology  and
neurobiology.

         Michael  Brownstein,  M.D.,  Ph.D.,  is Chief of the Laboratory of Cell
Biology at the National Institute of Mental Health. He is a recognized expert in
molecular pharmacology as it applies to the field of  neuroendocrinology,  where
he has defined many of the pharmaceutically important neurotransmitter receptors
and transporter systems.

         Roger D. Cone,  Ph.D., a Senior  Scientist at the Vollum  Institute for
Advanced Biomedical  Research,  is an international expert on the neuroendocrine
system,   with  particular   expertise  on  the  melanocortin   system  and  the
hypothalamic control of energy homeostasis. Dr. Cone is an editor of the journal
Endocrinology.

         Stephen M. Hedrick, Ph.D., is Professor and Chairman of Cell Biology at
the  University of  California,  San Diego.  Dr.  Hedrick is an expert in T cell
immunology and  co-discovered the first T cell receptor genes and identified the
regions  responsible  for  antigen  binding.  He is an editor for the Journal of
Immunology.

         Florian Holsboer,  M.D., Ph.D., is Director at the Max Planck Institute
fur  Psychiatrie.  Dr.  Holsboer  is an  international  expert  on the  role  of
glucocorticoids   and  neuropeptides,   particularly  CRF,  in  neuropsychiatric
disorders.  He  coordinates  the  efforts  of  several  hundred  scientists  and
clinicians  at the  Max  Planck  Institute,  a major  European  neuropsychiatric
institute.

         George   F.   Koob,   Ph.D.,   is  a  Member  of  the   Department   of
Neuropharmacology  at The Scripps Research Institute and an Adjunct Professor in
the  Departments  of Psychology  and Psychiatry at the University of California,
San Diego. Dr. Koob is an  internationally  recognized  behavioral  pharmacology
expert on the role of peptides in the central nervous system,  the neurochemical
basis of addiction and in the development of preclinical  behavioral  procedures
for the screening of anxiolytic and antidepressant drugs and memory enhancers.

         Charles B.  Nemeroff,  M.D.,  Ph.D.,  is Chairman and  Professor of the
Department of Psychiatry and Behavioral  Sciences at Emory University  School of
Medicine. Dr. Nemeroff is an internationally recognized expert on the effects of
neuropeptides on behavior and their relevance in clinically important conditions
such as  depression,  anxiety  and  schizophrenia,  and has  published  over 400
articles on this subject.



         Thomas  Roth,  Ph.D.,  is the Head of the  Division  of Sleep  Disorder
Research  at  the  Henry  Ford  Hospital  Research  Institute.  Dr.  Roth  is an
internationally  recognized expert in the field of sleep research.  His areas of
specialization are sleep homeostasis and neuropharmacology of sleep.

         Lawrence   J.   Steinman,    M.D.,   is   Chief   Scientific   Advisor,
Neuroimmunology  and a member of our Founding  Board of  Scientific  and Medical
Advisors and our Executive Committee.

         Wylie W. Vale, Ph.D., is Chief Scientific  Advisor,  Neuroendocrinology
and a member of our Founding  Board of Scientific  and Medical  Advisors and our
Executive Committee.

         Stanley J. Watson,  Jr., M.D.,  Ph.D., is Professor and Associate Chair
for Research in the  Department  of  Psychiatry  and  Co-Director  of the Mental
Health  Research  Institute  at the  University  of  Michigan.  Dr.  Watson is a
recognized  expert in  neuropeptides  and  their  receptors  and  their  role in
psychiatric diseases and behavior.  Dr. Watson is also a member of the Institute
of Medicine of the National Academy of Sciences.

         Each of the  members  of our  Scientific  Advisory  Board has  signed a
consulting agreement that contains confidentiality  provisions and restricts him
or her from competing with us for the term of the agreement.  Each member of our
Scientific  Advisory  Board  receives  either  a per  diem  consulting  fee or a
retainer fee. Each member also has received  Neurocrine  stock or stock options,
which vest over time. All members of our Scientific Advisory Board are full-time
employees  of a  university  or  research  institute  that has  regulations  and
policies,  which limit their ability to act as part-time consultants or in other
capacities  for any commercial  enterprise,  including  Neurocrine.  A change in
these  regulations or policies could adversely affect our relationship  with any
of our Scientific Advisory Board members.



ITEM 2.  PROPERTIES

         We lease approximately  93,000 square feet of space at our headquarters
in San  Diego,  California,  of  which  approximately  65% is  laboratory  space
dedicated to research and development. This facility was constructed in 1998 and
is under lease through  August 2013.  Our lease  payments are $208,000 per month
with  annual  increases  of 4% on  September  1st of each year.  We have  sublet
approximately  14,500  square  feet of this  building  to two  separate  tenants
through July 31, 2001 and September 30, 2001, respectively.

          We  believe  that  our  property  and  equipment  are  generally  well
maintained, in good operating condition and adequate for our current needs.


ITEM 3.  LEGAL PROCEEDINGS

         We are currently not subject to any material legal proceedings.


ITEM 4.  SUBMISSION OF MATTERS TO A VOTE OF SECURITY HOLDERS

         Not Applicable.





                                     PART II


ITEM 5.  MARKET FOR REGISTRANT'S COMMON EQUITY AND RELATED STOCKHOLDER MATTERS

         Our common stock has been traded on the Nasdaq  National  Market System
under the symbol NBIX since our initial public  offering on May 23, 1996.  Prior
to that time  there was no  established  public  trading  market  for our common
stock. The following table sets forth for the periods indicated the high and low
sale price for the common stock as reported by the Nasdaq National Market. These
prices do not include retail markups, markdowns or commissions.

                                                      High          Low
                                                      ----          ---
 Year Ended December 31, 1999
 1st Quarter ....................................... $ 7.50       $ 4.88
 2nd Quarter .......................................   5.88         4.00
 3rd Quarter .......................................   5.94         3.75
 4th Quarter .......................................  29.63         5.38
 Year Ended December 31, 2000
 1st Quarter .......................................  47.50        20.75
 2nd Quarter .......................................  39.75        13.94
 3rd Quarter .......................................  46.00        29.13
 4th Quarter .......................................  44.88        25.50

         As of February 28, 2001, there were  approximately  134 stockholders of
record of our common  stock.  We have not paid any cash  dividends on our common
stock  since  inception  and do not  anticipate  paying  cash  dividends  in the
foreseeable future.







ITEM 6.  SELECTED FINANCIAL DATA

         The following  selected  consolidated  financial data have been derived
from our audited consolidated  financial  statements.  The information set forth
below is not  necessarily  indicative  of the results of future  operations  and
should be read in  conjunction  with  "Management's  Discussion  and Analysis of
Financial  Condition and Results of Operations" and the  consolidated  financial
statements and notes thereto appearing elsewhere in this Form 10-K.




                                                2000     1999     1998 (1)   1997     1996
                                               ------   ------    -------   ------   ------
                                        (in thousands, except for earnings/(loss) per share data)
                                                                      
STATEMENT OF OPERATIONS DATA
Revenues
 Sponsored research and development ......  $  6,881  $ 12,171   $ 8,751   $14,985   $9,092
 Sponsored research and development
  from related party .....................         -       491     3,610         -        -
 Milestones and license fees .............     6,345     3,000     2,500    10,250    9,000
 Grant income and other revenues .........     1,362     1,129     1,176       909    1,124
                                              ------    ------    ------    ------   ------
 Total revenues ..........................    14,588    16,791    16,037    26,144   19,216

Operating expenses
 Research and development ................    40,227    29,169    21,803    18,758   12,569
 General and administrative ..............     9,962     7,476     6,594     5,664    3,697
 Write-off of acquired in-process
  research and development and licenses ..         -         -     4,910         -        -
                                              ------    ------    ------    ------   ------
 Total operating expenses ................    50,189    36,645    33,307    24,422   16,266

Income (loss) from operations ............   (35,601)  (19,854)  (17,270)    1,722    2,950

 Interest income, net ....................     6,048     2,851     4,000     3,931    2,598
 Other income (expense) ..................     1,047     1,066       504       818      574
 Equity in NPI net losses and other
  adjustments, net .......................         -      (885)   (7,188)   (1,130)       -
                                              ------    ------    ------    ------   ------
Net income (loss) before income taxes ....   (28,506)  (16,822)  (19,954)    5,341    6,122
Income taxes .............................       302         -         1       214      248
                                              ------    ------    ------    ------   ------
Net income (loss) ........................  $(28,808) $(16,822) $(19,955)  $ 5,127   $5,874
                                             ========  ========  ========   ======   ======

Earnings (loss) per share
 Basic ...................................  $  (1.30)  $ (0.88)  $ (1.10)   $ 0.30   $ 0.39
 Diluted .................................     (1.30)    (0.88)    (1.10)     0.28     0.36

Shares used in calculation of earnings
 (loss) per share
 Basic ...................................    22,124    19,072    18,141    16,930   14,971
 Diluted .................................    22,124    19,072    18,141    18,184   16,127

BALANCE SHEET DATA
Cash, cash equivalents and
  short-term investments .................   164,670    91,098    62,670    75,092   69,920
Working capital ..........................   154,556    86,168    60,064    69,362   68,023
Total assets .............................   185,962   109,222    80,529    91,903   77,957
Long-term debt and capital
 lease obligations .......................     2,283     2,139     2,247       722      847
Accumulated deficit ......................   (70,480)  (41,672)  (24,850)   (4,895) (10,022)
Total stockholders' equity ...............   163,208    96,354    71,958    83,152   72,767

- -----------------
<FN>

   (1)               Includes  results of operations  and financial  position of
                     Northwest  NeuroLogic,  Inc. from May 28, 1998, the date of
                     acquisition  (See  Note 8 of  the  Notes  to the  Financial
                     Statements).
</FN>




ITEM 7. MANAGEMENT'S DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND
                              RESULTS OF OPERATIONS

         The  following  Management's   Discussion  and  Analysis  of  Financial
Condition and Results of Operations section contains forward-looking  statements
which  involve  risks and  uncertainties,  pertaining  generally to the expected
continuation of our collaborative  agreements,  the receipt of research payments
thereunder,  the future achievement of various milestones in product development
and the receipt of payments  related thereto,  the potential  receipt of royalty
payments,  preclinical  testing and clinical trials of potential  products,  the
period  of time  that our  existing  capital  resources  will  meet our  funding
requirements, and our financial results and operations. Our actual results could
differ materially from those anticipated in these forward-looking  statements as
a result of various factors,  including those set forth below and those outlined
in the "Business" section included in Item 1.


Overview

         We incorporated in California in 1992 and reincorporated in Delaware in
1996.  Since  we were  founded,  we  have  been  engaged  in the  discovery  and
development  of novel  pharmaceutical  products  for  neurologic  and  endocrine
diseases  and  disorders.  Our product  candidates  address  some of the largest
pharmaceutical  markets in the world including  insomnia,  anxiety,  depression,
cancer and  diabetes.  To date, we have not generated any revenues from the sale
of  products,  and we do not expect to  generate  any  product  revenues  in the
foreseeable future. We have funded our operations  primarily through private and
public  offerings of our common stock and payments  received  under research and
development  agreements.  We are  developing a number of products with corporate
collaborators and will rely on existing and future collaborators to meet funding
requirements.  We expect to  generate  future  net  losses  in  anticipation  of
significant increases in operating expenses as products are advanced through the
various  stages of  clinical  development.  As of  December  31,  2000,  we have
incurred a  cumulative  deficit of $70.5  million and expect to incur  operating
losses in the future, which may be greater than losses in prior years.


Results of Operations

         Our  revenues for the year ended  December 31, 2000 were $14.6  million
compared with $16.8 million in 1999,  and $16.0 million in 1998.  The decline in
revenues  from  1999 to 2000  resulted  primarily  from  the  conclusion  of the
Novartis collaboration in January 2000 and the sponsored research portion of the
Eli Lilly & Company ("Eli Lilly") collaboration in October 1999. During 1999, we
received $6.8 million in revenues  under these  agreements,  in addition to $3.0
million  in  milestones  under the  Wyeth-Ayerst  Laboratories  ("Wyeth-Ayerst")
agreement.  The absence of these  revenues  during 2000 was partially  offset by
$7.1  million  in  revenues  received  from  Taisho  Pharmaceuticals  Co.,  Ltd.
("Taisho").  In addition,  revenues  received from Janssen  Pharmaceutica,  N.V.
("Janssen") were $3.0 million in 2000 compared to $2.4 million received in 1999.

         Revenues  for 1999 and 1998 were  similar  in total  but had  different
compositions  resulting from several significant events. During 1999, we entered
into a  collaborative  agreement  with  Wyeth-Ayerst  and  agreed to a  two-year
extension of our 1995 collaboration with Janssen.  The new agreements  generated
revenues of $8.4 million during 1999.  Non-recurring  revenues  recorded in 1998
included  $4.7 million in sponsored  development  and $2.3 million in milestones
received under the Novartis and Neuroscience Pharma, Inc. ("NPI") agreements. In
addition,  due to the  conclusion of the sponsored  research  portion of the Eli
Lilly  agreement in October 1999,  revenues  received from Eli Lilly during 1999
were $3.2 million compared to $4.1 million received in 1998.

         Research and  development  expenses  increased to $40.2 million  during
2000  compared  with  $29.2  million  during  1999 and  $21.8  million  in 1998.
Increased   expenses  reflect   advancement  of  our  drug  candidates   through
progressive   clinical  development  phases.  We  expect  to  incur  significant
increases in future periods as later phases of development  typically involve an
increase in the scope of studies,  the number of patients treated and the number
of scientific personnel required to manage the trials.

General and  administrative  expenses  increased  to $10.0  million  during 2000
compared  with $7.5  million  during  1999 and $6.6  million in 1998.  Increased
expenses  from 1999 to 2000  resulted  primarily  from $1.1  million in business



development  consulting  primarily  relating  to the Taisho  agreement  and $1.1
million of non-cash stock  compensation  charges  relating to the employee stock
purchase program and consultant stock options.  Increased  expenses from 1998 to
1999  resulted  primarily  from the  addition of  personnel  required to support
expanding  research and  development  activities.  We expect  these  expenses to
continue to rise in 2001 as we expand clinical studies.

         During 1998, we wrote-off acquired  in-process research and development
costs of $4.9  million.  This  amount  included  the  acquisition  of  Northwest
NeuroLogic,  Inc.  ("NNL")  and  the  in-licensing  of drug  candidates  for our
insomnia and malignant  glioma  programs.  Both of the in-licensed  programs are
currently under clinical development.

         Interest  income  increased to $6.3 million  during 2000  compared with
$3.1  million  during  1999 and $4.2  million  for 1998.  The  increase in 2000,
compared with 1999 and 1998,  primarily resulted from higher investment balances
achieved through offerings of our common stock. We completed a private placement
of 2.3  million  shares in December  1999,  resulting  in net  proceeds of $39.5
million.  In December  2000,  we sold 3.2 million  shares in a public  offering,
which  resulted in net  proceeds of $90.4  million.  Due to the increase in cash
reserves  generated  from these  transactions,  we  anticipate  an  increase  in
interest income during 2001.

         In December  1999, we sold our investment in NPI and recorded a gain of
$526,000.  Our proportionate  share of NPI operating losses during 1999 and 1998
were  $764,000  and $3.4  million,  respectively.  In  addition,  we  recorded a
write-down  in the  investment  value of $646,000  during 1999 and $3.8  million
during  1998  relating  to the  decline  in  cash  redemption  value  of the NPI
preferred shares.

         Other  income  consists  primarily  of sublease  income from  unrelated
parties.  The fluctuations in sublease income from year-to-year reflect facility
capacity in excess of our needs. Excess space is subleased until it is needed to
support  company  growth.  During 2001,  we expect  sublease  income to decrease
significantly  as increases in personnel will require more office and laboratory
space.

         Net loss for 2000 was $28.8 million, or $1.30 per share,  compared with
$16.8 million,  or $0.88 per share,  and $20.0 million,  or $1.10 per share, for
1999 and 1998, respectively. The increase in net loss primarily resulted from an
increase in scientific personnel and expanded clinical  development  activities.
We expect operating losses to increase for the foreseeable future as we continue
to expand our clinical development efforts.

         To date, our revenues have come  principally  from funded  research and
achievements of milestones under corporate collaborations. The nature and amount
of these revenues from period to period may lead to substantial  fluctuations in
the results of  year-to-year  revenues and  earnings.  Accordingly,  results and
earnings of one period are not predictive of future periods.


Liquidity and Capital Resources

         At December  31,  2000,  our cash,  cash  equivalents,  and  short-term
investments  totaled $164.7 million  compared with $91.1 million at December 31,
1999.  The  increase in cash  balances at December  31, 2000  resulted  from the
public offering of our common stock,  which generated net cash proceeds of $90.4
million.

         Net cash used by operating activities during fiscal year 2000 was $18.6
million  compared with $10.3 million  during 1999 and $10.7 million during 1998.
The increase in cash used in operations  during 2000 compared with 1999 and 1998
resulted primarily from the increase in clinical development  activities and the
addition of scientific personnel.

         Net cash used by investing activities during fiscal year 2000 was $75.7
million  compared to $21.2  million in 1999 and net cash  provided by  investing
activities of $4.7 million in 1998. The  fluctuations  in cash used and provided
resulted  primarily from the timing  differences  in the  investment  purchases,
sales,  maturities  and the  fluctuations  in our  portfolio  mix  between  cash
equivalents and short-term  investment holdings.  We expect similar fluctuations
to continue in future periods. Capital equipment purchases for 2001 are expected
to be approximately  $4.0 million and will be financed primarily through leasing
arrangements.

         Net cash provided by financing  activities  during fiscal year 2000 was
$94.1 million compared with $41.0 million and $1.9 million during 1999 and 1998,
respectively.  Cash provided  during 2000 includes $90.4 million of net proceeds
from the public offering of our common stock. Cash provided during 1999 includes



$39.5  million of net  proceeds  received  from the  private  sale of our common
stock.  Cash  provided  during  1998  resulted   primarily  from  capital  lease
financings.

         In July 2000, we licensed to Taisho the exclusive rights to develop and
commercialize  NBI-6024 in Europe and Asia.  In December  2000,  we expanded our
collaboration with Taisho,  providing Taisho the exclusive rights to develop and
commercialize our altered peptide ligand (APL) for diabetes in North America and
other countries outside of Europe and Asia. With the expanded agreement, we will
collaborate  in the  worldwide  clinical  development  of  NBI-6024  and we will
receive  funding for activities we conduct on behalf of the  collaboration.  The
worldwide collaboration is valued at up to $100 million, including all potential
licensing fees, purchase fees, milestones and development expenses. In addition,
we will receive  payments  based on any future  sales of  NBI-6024.  NBI-6024 is
currently in Phase I/II clinical trials,  with Phase II trials planned for 2001.
As of December  31, 2000,  we have  received  $2.0 million in option fees,  $4.0
million  in  license  fees,   $4.0  million  in  milestones,   and  $829,000  in
reimbursements  of  third-party  costs.  The license fees were  deferred and are
being recognized as revenues over the life of the agreement at $319,000 in 2000,
818,000 in each of the years 2001 through 2004 and $409,000 in 2005.

         In September  1999, we signed an amendment to our 1995  agreement  with
Janssen.  The amendment provides for a new sponsored research period designed to
identify new corticotropin-releasing  factor receptor antagonists, which will be
subject to the terms of the original  agreement  signed in 1995. The term of the
amendment is from April 1999 through February 2001. Under the agreement, we will
receive  $5.0  million in  sponsored  research  funding,  up to $3.5  million in
milestone  achievements and  reimbursement of all outside and third-party  costs
associated  with the project.  As of December 31, 2000,  we have  received  $4.6
million in  sponsored  research and $755,000 in  reimbursements  of  third-party
costs.

         In January 1999, we entered into an agreement  with  Wyeth-Ayerst,  the
pharmaceutical division of American Home Products, on the research,  development
and  commercialization  of  compounds,  which  modulate  excitatory  amino  acid
transporters for the treatment of neurodegenerative and psychiatric diseases.

         The   Wyeth-Ayerst   agreement   provides   for   sharing   proprietary
technologies,  funding for  research,  payments  for  milestones  reached,  plus
royalties on sales from products  resulting  from the  collaboration.  Under the
terms of the agreement,  we expect to receive three to five years of funding for
research and development as well as worldwide  royalties on commercial  sales of
products that result from the  collaboration.  Wyeth-Ayerst will also provide us
with access to chemical libraries for screening within the collaborative  field.
As of December 31, 2000,  we have  received  $6.0 million in sponsored  research
payments,  $3.0 million for the  achievement  of four  milestones and $50,000 in
license  fees,  which are being  deferred  and  recognized  over the life of the
agreement.

         During 1998, we expensed acquired  in-process  research and development
of $4.9 million.  These charges consisted of $4.2 million for the acquisition of
Northwest  NeuroLogic,  through which we received  licenses to the  melanocortin
receptor  and  excitatory  amino acid  transporters  programs,  and $710,000 for
licenses to insomnia and brain cancer  compounds.  We performed  scientific  due
diligence related to the acquired projects and because they were based on narrow
scientific hypotheses,  we concluded that none of these programs had alternative
future uses.

         The nature and  efforts  required to develop  the  acquired  in-process
research and development into  commercially  viable products include research to
identify a clinical candidate,  preclinical  development,  clinical testing, FDA
approval and commercialization.  This process may cost in excess of $100 million
and can take as long as 10 years to complete.  It is also important to note that
if a clinical candidate is identified, the further development of that candidate
can be halted or abandoned at any time due to a number of factors. These factors
include,  but are not limited  to,  funding  constraints,  safety or a change in
market demand.

         Because of our limited  financial  resources,  our  strategy to develop
some of our  programs  is to enter  into  collaborative  agreements  with  major
pharmaceutical  companies.  Through  these  collaborations,  we could  partially
recover our research costs through contract research and milestone revenues. The
collaborators would then be financially responsible for all clinical development
and commercialization costs.

         In May 1998,  when we acquired the in-process  research and development
programs from NNL, we estimated  the costs to identify a clinical  candidate and
provide minimal research support during the clinical  development stages for the
melanocortin  receptor program to be $15.4 million over an 8-year period.  Costs
to identify a clinical candidate and provide minimal research support during the



clinical  development  stages of the excitatory amino acid transporters  program
were  estimated at $22.4  million.  Estimated  revenues  from the  collaborative
arrangements were anticipated to reduce our net costs. The clinical  development
and commercialization costs were to be completely funded by the collaborator.

         During  fiscal  year  2001,  we  anticipate  that our  gross  costs for
continued  research on these programs will approximate $5 million.  Our research
efforts may not result in clinical candidates for either compound.  We intend to
collaborate  on the  melanocortin  receptor  technology.  We  would  expect  the
collaborator   to   then  be   responsible   for   the   clinical   development,
commercialization and funding. Our excitatory amino acid transporters program is
currently under a collaborative  agreement with Wyeth-Ayerst.  Consequently,  we
cannot  estimate the time or resources  they will commit to the  development  of
this program.

         Our insomnia and brain cancer compounds are both in the early stages of
clinical testing.  During 2001, we expect to spend  approximately $35 million on
additional  clinical  testing of the brain  cancer and  insomnia  compounds.  We
expect the clinical  testing of both compounds to continue for at least the next
two years, but our efforts may not result in commercially  viable  products.  If
our efforts  were  completely  successful  and we did not  collaborate  on these
compounds,  we estimate  that each compound  could cost an  additional  $50-$150
million and take up to five years to reach commercial viability.

         For  each  of our  programs,  we  periodically  assess  the  scientific
progress  and merits of the  programs to  determine  if  continued  research and
development is economically viable. Certain of our programs have been terminated
due to the lack of  scientific  progress  and  lack of  prospects  for  ultimate
commercialization.  Because of the  uncertainties  associated  with research and
development  of  these   programs,   we  may  not  be  successful  in  achieving
commercialization.  As such, the ultimate  timeline and costs to commercialize a
product cannot be accurately estimated.

         We believe that our existing capital resources,  together with interest
income and future payments due under our strategic alliances, will be sufficient
to satisfy our current and projected funding  requirements for at least the next
12 months.  However,  we cannot  guarantee  that  these  capital  resources  and
payments will be sufficient to conduct our research and development  programs as
planned. The amount and timing of expenditures will vary depending upon a number
of factors, including progress of our research and development programs.

         We will require additional funding to continue our research and product
development  programs,  to conduct  preclinical studies and clinical trials, for
operating expenses,  to pursue regulatory  approvals for our product candidates,
for the costs  involved  in  filing  and  prosecuting  patent  applications  and
enforcing or defending patent claims,  if any, the cost of product  in-licensing
and  any  possible  acquisitions,  and  we may  require  additional  funding  to
establish manufacturing and marketing capabilities in the future. We may seek to
access the public or private equity markets  whenever  conditions are favorable.
We may also  seek  additional  funding  through  strategic  alliances  and other
financing  mechanisms,  potentially  including  off-balance sheet financing.  We
cannot assure you that adequate funding will be available on terms acceptable to
us, if at all.  If  adequate  funds are not  available,  we may be  required  to
curtail  significantly  one or more of our research or  development  programs or
obtain funds through arrangements with collaborators or others. This may require
us to relinquish rights to certain of our technologies or product candidates.

         We expect to incur operating  losses over the next several years as our
research,  development,   preclinical  studies  and  clinical  trial  activities
increase.  To the extent  that we are unable to obtain  third-party  funding for
such expenses, we expect that increased expenses will result in increased losses
from  operations.  We cannot  assure you that we will  successfully  develop our
products under development or that our products, if successfully developed, will
generate revenues sufficient to enable us to earn a profit.


Interest Rate Risk

         We are exposed to interest rate risk on our short-term  investments and
on our long-term debt. The primary objective of our investment  activities is to
preserve   principal   while  at  the  same  time   maximizing   yields  without
significantly  increasing  risk. To achieve this objective,  we invest in highly
liquid and high quality  government and other debt  securities.  To minimize our
exposure  due to  adverse  shifts in  interest  rates,  we invest in  short-term



securities with  maturities of less than 44 months.  If a 10% change in interest
rates were to have occurred on December 31, 2000, this change would not have had
a material effect on the fair value of our investment portfolio as of that date.
Due to the short holding period of our investments, we have concluded that we do
not have a material financial market risk exposure.

         Interest risk  exposure on long-term  debt relates to our note payable,
which bears a floating interest rate of prime plus one quarter percent (9.75% at
December 31, 2000,  8.75% at December 31, 1999 and 8.00% at December 31,  1998).
At December 31, 2000, 1999 and 1998, the note balance was $311,000, $461,000 and
$610,000,  respectively.  This note is  payable  in equal  monthly  installments
through  January  2003.  Based on the  balance of our  long-term  debt,  we have
concluded that we do not have a material financial market risk exposure.


Cautionary Note on Forward-Looking Statements

         Our business is subject to significant risks, including but not limited
to, the risks inherent in our research and development activities, including the
successful  continuation  of  our  strategic   collaborations,   the  successful
completion of clinical trials,  the lengthy,  expensive and uncertain process of
seeking regulatory approvals,  uncertainties  associated both with the potential
infringement of patents and other intellectual property rights of third parties,
and  with   obtaining  and   enforcing  our  own  patents  and  patent   rights,
uncertainties   regarding   government   reforms  and  of  product  pricing  and
reimbursement  levels,  technological  change  and  competition,   manufacturing
uncertainties  and dependence on third parties.  Even if our product  candidates
appear promising at an early stage of development, they may not reach the market
for numerous reasons.  Such reasons include the  possibilities  that the product
will be  ineffective  or unsafe  during  clinical  trials,  will fail to receive
necessary  regulatory  approvals,  will be difficult to  manufacture  on a large
scale,   will  be   uneconomical   to   market   or  will  be   precluded   from
commercialization  by proprietary rights of third parties.  For more information
about the risks we face, see "Risk Factors" included in Part I of this report.


New Accounting Pronouncements

         In December  1999,  the SEC issued Staff  Accounting  Bulletin No. 101,
"Revenue  Recognition  in  Financial  Statements"  (SAB 101).  SAB 101  provides
guidance  in  applying  generally  accepted  accounting  principles  to  revenue
recognition in financial statements,  including the recognition of nonrefundable
up-front  fees  received  in  conjunction   with  a  research  and   development
arrangement.  The adoption of this pronouncement was required effective with the
fourth quarter of 2000.

         As required by the adoption, we reviewed all up-front payments, license
fees and milestones  received in the current and prior years.  Up-front payments
have been received for program cost reimbursements incurred during a negotiation
period. License fees are received in exchange for a grant to use our proprietary
technologies  on an as-is basis,  for the term of the  collaborative  agreement.
Milestones are received for specific scientific  achievements  determined at the
beginning of the collaboration.  These achievements are remote and unpredictable
at the onset of the  collaboration  and are based on the  success of  scientific
efforts.

         Based on that review,  we determined  that $4.2 million of license fees
received  during 2000 were  subject to the  adoption of SAB 101.  All other fees
received  relate  to  agreements   under  which  the  research  portion  of  the
collaboration  has  been  completed  or  the  agreements  have  been  terminated
entirely.  In accordance with Accounting  Principles Board (APB) Opinion No. 20,
the adoption of SAB 101 was recognized by including the cumulative effect of the
change in accounting  principle in the net loss for the fourth  quarter of 2000.
Our  otherwise  reported  net loss  for the year  ended  December  31,  2000 was
increased  by  approximately  $3.8  million.  These  license fee  revenues  were
deferred and will be amortized as income at $915,000 in 2001,  $835,000 in 2002,
$828,000 in 2003, $818,000 in 2004 and $409,000 in 2005.

         In March 2000,  the  Financial  Accounting  Standards  Board,  or FASB,
released  Interpretation No. 44, "Accounting for Certain Transactions  involving
Stock  Compensation,  an  interpretation  of APB Opinion No. 25," (FIN 44) which
provides   clarification   of  Opinion  25  for  certain   issues  such  as  the
determination of who is an employee, the criteria for determining whether a plan
qualifies as a  non-compensatory  plan,  the  accounting  consequence of various



modifications  to the terms of a previously fixed stock option or award, and the
accounting  for  an  exchange  of  stock  compensation   awards  in  a  business
combination. We believe that our practices are in conformity with this guidance,
and therefore FIN 44 had no impact on our financial statements.

         In June  1998,  the  FASB  issued  Statement  of  Financial  Accounting
Standards  (SFAS) No.  133,"Accounting  for Derivative  Instruments  and Hedging
Activities."  We expect to adopt the new  Statement  effective  January 1, 2001.
This statement requires the recognition of all derivative  instruments as either
assets or liabilities in the statement of financial position and the measurement
of those  instruments at fair value. The Company does not expect the adoption of
this  statement  to have a  material  impact on its  results  of  operations  or
financial position.

         In  September  2000,  the FASB  issued SFAS No.  140,  "Accounting  for
Transfers and Servicing of Financial Assets and Extinguishments of Liabilities."
SFAS 140 provides accounting and reporting standards for transfers and servicing
of financial  assets and  extinguishments  of  liabilities  and is effective for
transfers and servicing of financial assets and  extinguishments  of liabilities
occurring after March 31, 2001. The adoption of SFAS 140 is not expected to have
a material impact on our financial statements.


ITEM 7A.  QUANTITATIVE AND QUALITATIVE DISCLOSURES ABOUT MARKET RISK

         Quantitative and Qualitative Disclosures about Market Risk is contained
in Item 7, Management Discussion and Analysis--Interest Rate Risk.


ITEM 8.  FINANCIAL STATEMENTS AND SUPPLEMENTARY DATA

         See the list of the Company's Financial Statements filed with this Form
10-K under Item 14 below.


ITEM 9.   CHANGES IN AND DISAGREEMENTS WITH ACCOUNTANTS ON ACCOUNTING AND
          FINANCIAL DISCLOSURE

         Not Applicable.




                                    PART III


ITEM 10.  EXECUTIVE OFFICERS AND DIRECTORS OF THE REGISTRANT

         Information  required by this item will be contained  in the  Company's
Notice of 2001 Annual Meeting of Stockholders and Proxy  Statement,  pursuant to
Regulation 14A, to be filed with the Securities and Exchange  Commission  within
120 days of December  31,  2000.  Such  information  is  incorporated  herein by
reference.


ITEM 11.  EXECUTIVE COMPENSATION

         Information  required by this item will be contained  in the  Company's
Notice of 2001 Annual Meeting of Stockholders and Proxy  Statement,  pursuant to
Regulation 14A, to be filed with the Securities and Exchange  Commission  within
120 days of December  31,  2000.  Such  information  is  incorporated  herein by
reference.


ITEM 12.  SECURITY OWNERSHIP OF CERTAIN BENEFICIAL OWNERS AND MANAGEMENT

         Information  required by this item will be contained  in the  Company's
Notice of 2001 Annual Meeting of Stockholders and Proxy  Statement,  pursuant to
Regulation 14A, to be filed with the Securities and Exchange  Commission  within
120 days of December  31,  2000.  Such  information  is  incorporated  herein by
reference.


ITEM 13.  CERTAIN RELATIONSHIPS AND RELATED TRANSACTIONS

         Information  required by this item will be contained  in the  Company's
Notice of 2001 Annual Meeting of Stockholders and Proxy  Statement,  pursuant to
Regulation 14A, to be filed with the Securities and Exchange  Commission  within
120 days of December  31,  2000.  Such  information  is  incorporated  herein by
reference.



                                     PART IV


ITEM 14.  EXHIBITS, FINANCIAL STATEMENT SCHEDULES AND REPORTS ON FORM 8-K

(a)      Documents filed as part of this report

         1.       List  of  Financial   Statements.   The  following   financial
                  statements of Neurocrine Biosciences, Inc. and Report of Ernst
                  &  Young  LLP,  Independent  Auditors,  are  included  in this
                  report:  Report  of Ernst & Young  LLP,  Independent  Auditors
                  Consolidated  Balance  Sheet as of December  31, 2000 and 1999
                  Consolidated  Statement  of  Operations  for the  years  ended
                  December 31,  2000,  1999 and 1998  Consolidated  Statement of
                  Stockholders'  Equity for the years ended  December  31, 2000,
                  1999 and 1998  Consolidated  Statement  of Cash  Flows for the
                  years  ended  December  31,  2000,  1999 and 1998 Notes to the
                  Consolidated Financial Statements (includes unaudited Selected
                  Quarterly Financial Data)

         2.       List of all Financial Statement  schedules.  All schedules are
                  omitted  because  they  are  not  applicable  or the  required
                  information is shown in the Consolidated  Financial Statements
                  or notes thereto.

         3.       List of Exhibits  required by Item 601 of Regulation  S-K. See
                  part (c) below.



(b)      Reports on Form 8-K.  Current  Reports filed pursuant to Sections 13(a)
         or 15(d) of the Exchange  Act on Forms 8-K dated  December 14, 2000 and
         December 15, 2000.

         1.       The Registrant  filed a Current Report on Form 8-K dated April
                  6, 2000 to report pursuant to Item 5 (Other Events) and Item 7
                  (Final  Statements,   Pro  Forma  Financial   Information  and
                  Exhibits)  announcing Janssen  Pharmaceutica's  replacement of
                  R121919 with a back-up compound

         2.       The  Registrant  filed a  Current  Report  on Form  *-K  dated
                  December 14, 2000 to report  pursuant to Item 5 (Other Events)
                  and Item 7 (Final Statements,  Pro Forma Financial Information
                  and  Exhibits)  the  incorporation  by  reference of its final
                  prospectus  dated December 5, 2000. 3. The Registrant  filed a
                  Current  Report on Form 8-K dated  December 15, 2000 to report
                  pursuant  to  Item  5  (Other  Events)  the  expansion  of its
                  collaboration with Taisho Pharmaceuticals Co., LTD.

(c)      Exhibits.  The following exhibits are filed as part of, or incorporated
         by reference into, this report:

Exhibit
Number           Description
- --------------------------------------------------------------------------------

2.1      Agreement  and  Plan  of  Reorganization  dated  May 1,  1998,  between
         Northwest  NeuroLogic,   Inc.,  NBI  Acquisition  Corporation  and  the
         Registrant (7)

2.2      Form of Warrant  pursuant to the Agreement  and Plan of  Reorganization
         dated May 1, 1998 (7)

3.1      Restated Certificate of Incorporation (1)

3.2      Bylaws (1)

3.3      Certificate of Amendment of Bylaws (1)

4.1      Form of Common Stock Certificate (1)

4.2      Form of warrant issued to existing warrant holders (1)

4.3      Information and Registration Rights Agreement dated September 15, 1992,
         as amended (1)

4.4*     Registration  Rights  Agreement  dated May 28,  1998,  between  certain
         investors and the Registrant (7)

4.5      Amended and Restated Rights Agreement by and between the Registrant and
         American Stock Transfer & Trust Company,  as Rights Agent,  dated as of
         July 19, 1999 (9)

4.6      Stock Purchase  Agreement  dated December 20 through 23, 1999,  between
         Neurocrine  Biosciences,  Inc. and each of the Purchasers named therein
         (11)

10.1     Purchase and Sale Agreement and Escrow Instructions  between MS Vickers
         II, LLC and the Registrant dated February 13, 1997 (3)

10.2     1992 Incentive Stock Plan, as amended (10)

10.3     1996 Employee Stock Purchase Plan (1)

10.4     1996 Director Stock Option Plan and form of stock option agreement (1)

10.5     Form of Director and Officer Indemnification Agreement (1)

10.6     Employment  Agreement  dated March 1, 1997,  between the Registrant and
         Gary A. Lyons, as amended May 24, 2000 (4) (13)

10.7     Employment  Agreement  dated March 1, 1997,  between the Registrant and
         Paul W. Hawran, as amended May 24, 2000 (4) (13)

10.8     Consulting  Agreement dated September 25, 1992,  between the Registrant
         and Wylie A. Vale, Ph.D. (1)

10.9     Consulting  Agreement effective January 1, 1992, between the Registrant
         and Lawrence J. Steinman, MD (1)

10.10    License  Agreement  dated  July  17,  1992,  by and  between  The  Salk
         Institute for Biological Studies and the Registrant (1)

10.11    License  Agreement  dated  November 16,  1993,  by and between The Salk
         Institute for Biological Studies and the Registrant (1)

10.12    License  Agreement  dated October 19, 1992, by and between the Board of
         Trustees of the Leland  Stanford  Junior  University and the Registrant
         (1)

10.13    Agreement  dated  January 1, 1995,  by and between the  Registrant  and
         Janssen Pharmaceutica, N.V. (1)

10.14*   Research and License  Agreement  dated  October 15,  1996,  between the
         Registrant and Eli Lilly and Company (2)



10.15*   Lease between Science Park Center LLC and the Registrant dated July 31,
         1997 (5)

10.16*   Option  Agreement  between  Science Park Center LLC  (Optionor) and the
         Registrant dated July 31, 1997 (Optionee) (5)

10.17*   Construction  Loan Agreement Science Park Center LLC and the Registrant
         dated July 31, 1997 (5)

10.18    Secured  Promissory  Note  Science  Park Center LLC and the  Registrant
         dated July 31, 1997 (5)

10.19*   Operating   Agreement   for  Science  Park  Center  LLC  between  Nexus
         Properties, Inc. and the Registrant dated July 31, 1997 (5)

10.20    Form of incentive stock option agreement and nonstatutory  stock option
         agreement for use in connection with 1992 Incentive Stock Plan (1)

10.21*   Patent  License  Agreement  dated May 7,  1998,  between  the US Public
         Health Service and the Registrant (7)

10.22*   Patent License  Agreement  dated April 28, 1998,  between and among Ira
         Pastan, David Fitzgerald and the Registrant (7)

10.23*   Sub-License  and  Development  Agreement  dated June 30,  1998,  by and
         between DOV Pharmaceutical, Inc. and the Registrant (7)

10.24*   Warrant Agreement dated June 30, 1998, between DOV Pharmaceutical, Inc.
         and the Registrant (7)

10.25*   Warrant  Agreement  dated June 30, 1998,  between Jeff Margolis and the
         Registrant (7)

10.26*   Warrant  Agreement  dated June 30, 1998,  between  Stephen Ross and the
         Registrant (7)

10.27*   Collaboration  and  License  Agreement  dated  January 1, 1999,  by and
         between   American  Home  Products   Corporation   acting  through  its
         Wyeth-Ayerst Laboratories Division and the Registrant (8)

10.28*   Employment  Agreement dated January 1, 1999, between the Registrant and
         Margaret Valeur-Jensen, as amended May 24, 2000 (8) (13)

10.29*   Employment Agreement dated February 9, 1998, between the Registrant and
         Bruce Campbell, as amended May 24, 2000 (8) (13)

10.30    Amended 1992  Incentive  Stock Plan,  as amended May 27, 1997,  May 27,
         1998 and May 21, 1999 (8)

10.31*   Agreement  by  and  among  Dupont  Pharmaceuticals   Company,   Janssen
         Pharmaceutica,  N.V. and Neurocrine  Biosciences,  Inc. dated September
         28, 1999 (10)

10.32*   Amendment Number One to the Agreement between  Neurocrine  Biosciences,
         Inc. and Janssen Pharmaceutica, N.V. dated September 24, 1999 (10)

10.33*   License Agreement between the Registrant and Taisho Pharmaceutical Co.,
         Ltd. dated July 21, 2000 (13)

10.34**  Amendment  No.  1 dated  November  30,  2000 to the  License  Agreement
         between the Registrant and Taisho  Pharmaceutical  Co., Ltd. dated July
         21, 2000

10.35    2001 Stock Option Plan (14)

21.1     Subsidiaries of the Company

23.1     Consent of Ernst & Young LLP, Independent Auditors

24.1     Power of Attorney (see page 34)

- -----------

         (1)      Incorporated  by  reference  to  the  Company's   Registration
                  Statement on Form S-1 (Registration No. 333-03172)

         (2)      Incorporated by reference to the Company's Report on Form 10-K
                  for the fiscal year ended December 31, 1996

         (3)      Incorporated by reference to the Company's  amended  Quarterly
                  Report on Form 10-Q filed on August 15, 1997

         (4)      Incorporated by reference to the Company's Quarterly Report on
                  Form 10-Q filed on August 14, 1997

         (5)      Incorporated by reference to the Company's Quarterly Report on
                  Form 10-Q filed on November 14, 1997

         (6)      Incorporated by reference to the Company's  Report on Form 8-K
                  filed on March 13, 1998

         (7)      Incorporated by reference to the Company's Quarterly Report on
                  Form 10-Q filed on November 16, 1998

         (8)      Incorporated by reference to the Company's Report on Form 10-K
                  for the fiscal year ended December 31, 1998

         (9)      Incorporated by reference to the Company's Quarterly Report on
                  Form 10-Q filed on August 11, 1999

         (10)     Incorporated by reference to the Company's Quarterly Report on
                  Form 10-Q filed on November 12, 1999

         (11)     Incorporated by reference to the Company's  Report on Form S-3
                  filed on January 20, 2000

         (12)     Incorporated  by  reference to the  Company's  Report on Proxy
                  filed on April 27, 2000



         (13)     Incorporated by reference to the Company's Quarterly Report on
                  Form 10-Q filed on August 11, 2000

         (14)     Incorporated  by  reference  to  the  Company's   Registration
                  Statement  on Form S-8  filed  March 15,  2001 *  Confidential
                  treatment has been granted with respect to certain portions of
                  the exhibit ** Confidential  treatment has been requested with
                  respect to certain portions of the exhibit

(d)      Financial Statement Schedules. See Item 14 (a)(2) above.


                                   SIGNATURES

         Pursuant to the  requirements  of Section 13 or 15(d) of the Securities
Exchange Act of 1934, the Registrant has duly caused this report to be signed on
its behalf by the undersigned, thereunto duly authorized.

                                    NEUROCRINE BIOSCIENCES, INC.
                                      A Delaware Corporation


                                    By: /s/Gary A. Lyons
                                        ----------------
                                        Gary A. Lyons
                                        President and Chief Executive Officer

                                    Date:  March 29, 2001





                                POWER OF ATTORNEY

         KNOW ALL PERSONS BY THESE  PRESENTS,  that each person whose  signature
appears below  constitutes  and appoints Gary A. Lyons and Paul Hawran,  jointly
and severally his  attorneys-in-fact,  each with the power of substitution,  for
him in any and all  capacities,  to sign any  amendment  to this  Report on Form
10-K,  and to file the  same,  with  exhibits  thereto  and other  documents  in
connection  therewith,  with the  Securities  and  Exchange  Commission,  hereby
ratifying  and  confirming  all  that  each  of said  attorneys-in-fact,  or his
substitute or substitutes, may or cause to be done by virtue hereof.

         Pursuant to the  requirements  of the Securities  Exchange Act of 1934,
this report has been signed by the following persons on behalf of the Registrant
and in the capacities and on the dates indicated:

Signature                              Title                           Date
- ---------                              -----                           ----

/s/ Gary A. Lyons              President, Chief Executive        March 29, 2001
- ----------------------------      Officer and Director
Gary A. Lyons                (Principal Executive Officer)


/s/ Paul W. Hawran             Chief Financial Officer           March 29, 2001
- ----------------------------   (Principal Financial
Paul W. Hawran                 and Accounting Officer)


/s/ Joseph A. Mollica          Chairman of the                   March 29, 2001
- ----------------------------   Board of Directors
Joseph A. Mollica.


/s/ Richard F. Pops            Director                          March 29, 2001
- ----------------------------
Richard F. Pops


/s/ Stephen A. Sherwin         Director                          March 29, 2001
- ----------------------------
Stephen A. Sherwin


/s/ Lawrence J. Steinman       Director                          March 29, 2001
- ----------------------------
Lawrence J. Steinman


/s/ Wylie W. Vale              Director                          March 29, 2001
- ----------------------------
Wylie W. Vale






                          NEUROCRINE BIOSCIENCES, INC.
                 INDEX TO THE CONSOLIDATED FINANCIAL STATEMENTS




                                                                       Page

Report of Ernst & Young LLP, Independent Auditors ....................  F-2

Consolidated Balance Sheet ...........................................  F-3

Consolidated Statement of Operations .................................  F-4

Consolidated Statement of Stockholders' Equity .......................  F-5

Consolidated Statement of Cash Flows .................................  F-6

Notes to the Consolidated Financial Statements .......................  F-7




                REPORT OF ERNST & YOUNG LLP, INDEPENDENT AUDITORS



The Board of Directors and Stockholders
Neurocrine Biosciences, Inc.

We have  audited  the  accompanying  consolidated  balance  sheet of  Neurocrine
Biosciences, Inc. as of December 31, 2000 and 1999, and the related consolidated
statements of operations,  stockholders'  equity, and cash flows for each of the
three years in the period ended December 31, 2000.  These  financial  statements
are the  responsibility of the Company's  management.  Our  responsibility is to
express an opinion on these financial statements based on our audits.

We conducted our audits in accordance with auditing standards generally accepted
in the United States. Those standards require that we plan and perform the audit
to obtain reasonable  assurance about whether the financial  statements are free
of material misstatement. An audit includes examining, on a test basis, evidence
supporting  the amounts and  disclosures in the financial  statements.  An audit
also includes assessing the accounting principles used and significant estimates
made by  management,  as well as  evaluating  the  overall  financial  statement
presentation.  We believe  that our audits  provide a  reasonable  basis for our
opinion.

In our opinion,  the financial  statements  referred to above present fairly, in
all  material  respects,  the  consolidated  financial  position  of  Neurocrine
Biosciences, Inc. at December 31, 2000 and 1999, and the consolidated results of
its  operations  and its cash  flows for each of the three  years in the  period
ended  December 31, 2000, in conformity  with  accounting  principles  generally
accepted in the United States.

As discussed in Note 1 to the consolidated  financial  statements,  in 2000, the
Company changed its method of revenue recognition.



                                                  /s/ ERNST & YOUNG LLP
                                                  ---------------------
                                                  ERNST & YOUNG LLP

San Diego, California
January 29, 2001






                          NEUROCRINE BIOSCIENCES, INC.
                           Consolidated Balance Sheet
                                 (in thousands)

                                                                December 31,
                                                           ---------------------
                                                              2000       1999
                                                           ---------   ---------
                                   ASSETS
Current assets:
    Cash and cash equivalents ............................  $ 21,078   $ 21,265
    Short-term investments, available-for-sale ...........   143,592     69,833
    Receivables under collaborative agreements ...........     5,974      1,458
    Other current assets .................................     1,761      2,257
                                                           ---------   ---------
       Total current assets ..............................   172,405     94,813

    Property and equipment, net ..........................    11,300     11,181
    Licensed technology and patent applications costs, net       362        615
    Other assets .........................................     1,895      2,613
                                                           ---------   ---------
       Total assets ......................................  $185,962   $109,222
                                                           =========   =========

                      LIABILITIES AND STOCKHOLDERS' EQUITY
Current liabilities:
    Accounts payable ..................................... $   1,065   $  2,447
    Accrued liabilities ..................................    11,135      5,069
    Deferred revenues ....................................     1,172        155
    Current portion of long-term debt ....................       149        149
    Current portion of capital lease obligations .........     1,438        825
                                                           ---------   ---------
       Total current liabilities .........................    14,959      8,645

    Long-term debt, net of current portion ...............       162        312
    Capital lease obligations, net of current portion ....     2,121      1,827
    Deferred rent ........................................     1,646      1,005
    Deferred revenues ....................................     2,890          -
    Other liabilities ....................................       976      1,079
                                                           ---------   ---------
       Total liabilities .................................    22,754     12,868

Commitments and contingencies (See Note 6) ...............        -          -

Stockholders' equity:
    Preferred Stock, $0.001 par value; 5,000,000 shares
       authorized; no shares issued and outstanding ......        -          -
    Common Stock, $0.001 par value; 50,000,000 shares
       authorized; issued and outstanding shares were
       25,314,470 in 2000 and 21,608,011 in 1999 .........       25          22
    Additional paid in capital ...........................  233,565     138,798
    Deferred compensation ................................      (59)       (411)
    Stockholder notes ....................................     (104)       (119)
    Accumulated other comprehensive (loss) income ........       261       (264)
    Accumulated deficit ..................................  (70,480)    (41,672)
                                                           ---------   ---------
       Total stockholders' equity ........................  163,208      96,354
                                                           ---------   ---------
       Total liabilities and stockholders' equity ........ $185,962    $109,222
                                                           =========   =========



                             See accompanying notes.





                          NEUROCRINE BIOSCIENCES, INC.
                      Consolidated Statement of Operations
                                 (in thousands)

                                                     Year-ended December 31,
                                                  ----------------------------
                                                    2000      1999      1998
                                                  -------   -------    -------
Revenues:
 Sponsored research and development ...........  $  6,881  $ 12,171   $  8,751
 Sponsored research and development
  from related party ..........................         -       491      3,610
 Milestones and license fees ..................     6,345     3,000      2,500
 Grant income and other revenues ..............     1,362     1,129      1,176
                                                  -------   -------    -------
    Total revenues ............................    14,588    16,791     16,037

Operating expenses:
  Research and development ....................    40,227    29,169     21,803
  General and administrative ..................     9,962     7,476      6,594
  Write-off of acquired in-process research
   and development and licenses ...............         -         -      4,910
                                                  -------   -------    -------
     Total operating expenses .................    50,189    36,645     33,307

Income (loss) from operations .................   (35,601)  (19,854)   (17,270)

Other income and expenses:
 Interest income ..............................     6,276     3,082      4,151
 Interest expense .............................      (228)     (231)      (151)
 Equity in NPI losses and
  other adjustments, net ......................         -      (885)    (7,188)
 Other income .................................     1,047     1,066        504
                                                  -------   -------    -------
Income (loss) before taxes ....................   (28,506)  (16,822)   (19,954)
Income taxes ..................................       302         -          1
                                                  -------   -------    -------
Net income (loss) .............................  $(28,808) $(16,822)  $(19,955)
                                                  =======   =======    =======
Earnings (loss) per common share:
 Basic and diluted ............................  $  (1.30) $  (0.88)  $  (1.10)

Shares used in the calculation of
 earnings (loss) per common share:
 Basic and diluted ............................    22,124    19,072     18,141



                             See accompanying notes.




                          NEUROCRINE BIOSCIENCES, INC.
                 Consolidated Statement of Stockholders' Equity
                                 (in thousands)




                                                                                                      Notes
                                                        Common Stock     Additional                Receivable
                                                    -------------------   Paid In     Unearned        from
                                                      Shares    Amount    Capital   Compensation   Stockholders
                                                    --------------------------------------------------------------
                                                                                       
     BALANCE AT DECEMBER 31, 1997 .................   17,687      $18    $ 88,586     $ (439)         $(120)
Net loss ..........................................        -        -           -          -              -
Unrealized gain on short-term investments .........        -        -           -          -              -

Comprehensive loss ................................        -        -           -          -              -

Issuance of common stock for warrants .............       60        -         142          -              -
Issuance of common stock for option exercises .....       81        -         286          -              -
Issuance of common stock pursuant to the
  Employee Stock Purchase Plan ....................       30        -         205          -              -
Issuance of common stock in exchange for
  NPI Preferred Stock .............................      679        1       3,854          -              -
Issuance of common stock for NNL Acquisition ......      392        -       4,032          -              -
Issuance of common stock for milestone achievement         2        -          17          -              -
Payments received on stockholder notes ............        -        -           -          -              1
Amortization of deferred compensation, net ........        -        -         (58)       252              -
                                                     -------------------------------------------------------------
     BALANCE AT DECEMBER 31, 1998 .................   18,931       19      97,064       (187)          (119)

Net loss ..........................................        -        -           -          -              -
Unrealized gain on short-term investments .........        -        -           -          -              -

Comprehensive loss ................................        -        -           -          -              -
Issuance of common stock for option exercises .....      307        -       1,507          -              -
Issuance of common stock pursuant to the Employee
Stock  Purchase Plan ..............................       42        -         213          -              -
Issuance of common stock, net of offering costs ...    2,328        3      39,293          -              -
Amortization of deferred compensation, net ........        -        -         721       (224)             -
                                                     -------------------------------------------------------------
     BALANCE AT DECEMBER 31, 1999 .................   21,608       22     138,798       (411)          (119)

Net loss ..........................................        -        -           -          -              -
Unrealized gain on short-term investments .........        -        -           -          -              -

Comprehensive loss ................................        -        -           -          -              -
Issuance of common stock for warrants .............       23                    -          -              -
Issuance of common stock for stock purchase agreement      6        -           1          -              -
Issuance of common stock for option exercises .....      354        -       2,328          -              -
Issuance of common stock pursuant to the Employee
Stock  Purchase Plan ..............................       98        -       1,339          -              -
Issuance of common stock, net of offering costs ...    3,225        3      90,353          -              -
Payments received on stockholder notes ............        -        -           -          -             15
Reversal of accrued 12/99 private placement costs .        -        -         182          -              -
Amortization of deferred compensation, net ........        -        -         564        352              -
                                                     -------------------------------------------------------------
     BALANCE AT DECEMBER 31, 2000 .................   25,314      $25    $233,565     $  (59)         $(104)
                                                     =============================================================


                          NEUROCRINE BIOSCIENCES, INC.
                 Consolidated Statement of Stockholders' Equity
                                 (in thousands, continued)



                                                     Accumulated
                                                        Other                      Total
                                                    Comprehensive  Accumulated Stockholders'
                                                    Income (Loss)    Deficit      Equity
                                                    -------------------------------------
                                                                          
     BALANCE AT DECEMBER 31, 1997 .................    $   2       $ (4,895)       $ 83,152
Net loss ..........................................        -        (19,955)        (19,955)
Unrealized gain on short-term investments .........       29              -              29
                                                                                    -------
Comprehensive loss ................................        -              -         (19,926)

Issuance of common stock for warrants .............        -              -             142
Issuance of common stock for option exercises .....        -              -             286
Issuance of common stock pursuant to the
  Employee Stock Purchase Plan ....................        -              -             205
Issuance of common stock in exchange for
  NPI Preferred Stock .............................        -              -           3,855
Issuance of common stock for NNL Acquisition ......        -              -           4,032
Issuance of common stock for milestone achievement         -              -              17
Payments received on stockholder notes ............        -              -               1
Amortization of deferred compensation, net ........        -              -             194
                                                    ---------------------------------------
     BALANCE AT DECEMBER 31, 1998 .................       31        (24,850)         71,958

Net loss ..........................................        -        (16,822)        (16,822)
Unrealized gain on short-term investments .........     (295)             -            (295)
                                                                                    -------
Comprehensive loss ................................        -              -         (17,117)
Issuance of common stock for option exercises .....        -              -           1,507
Issuance of common stock pursuant to the Employee
Stock  Purchase Plan ..............................        -              -             213
Issuance of common stock, net of offering costs ...                                  39,296
Amortization of deferred compensation, net ........        -              -             497
                                                    ---------------------------------------
     BALANCE AT DECEMBER 31, 1999 .................     (264)       (41,672)         96,354

Net loss ..........................................        -        (28,808)        (28,808)
Unrealized gain on short-term investments .........      525              -             525
                                                                                    -------
Comprehensive loss ................................        -              -          68,071
Issuance of common stock for warrants .............        -              -
Issuance of common stock for stock purchase agreement      -              -               1
Issuance of common stock for option exercises .....        -              -           2,328
Issuance of common stock pursuant to the Employee
Stock  Purchase Plan ..............................        -              -           1,339
Issuance of common stock, net of offering costs                                      90,356
Payments received on stockholder notes ............        -              -              15
Reversal of accrued 12/99 private placement costs .        -              -             182
Amortization of deferred compensation, net ........        -              -             916
                                                    ---------------------------------------
     BALANCE AT DECEMBER 31, 2000 .................    $ 261       $(70,480)       $163,208
                                                    =======================================


                             See accompanying notes.




                          NEUROCRINE BIOSCIENCES, INC.
                      Consolidated Statement of Cash Flows
                                 (in thousands)



                                                                 Twelve Months Ended December 31,
                                                               ----------------------------------
                                                                  2000        1999        1998
                                                            ----------  ----------  ----------
CASH FLOW FROM OPERATING ACTIVITIES                                               
Net (loss) income ...........................................  $(28,808)    $(16,822)   $(19,955)
Adjustments to reconcile net income (loss) to net cash
  Provided by (used in) operating activities:
      Acquisition of Northwest NeuroLogic for Common Stock ..         -            -       4,200
      Equity in NPI losses and other adjustments, net .......         -          885       7,188
      Depreciation and amortization .........................     2,198        2,066       1,720
      Loss on abandonment of assets .........................        80          133         460
      Gain on sale of equipment .............................         -            -         (15)
      Deferred revenues .....................................     3,907          (14)     (1,750)
      Deferred rent .........................................       868          748        (402)
      Compensation expenses recognized for stock options ....     2,677          497         194
      Change in operating assets and liabilities,
        net of acquired business:
          Accounts receivable and other current assets ......    (4,020)        (752)     (2,898)
          Other non-current assets ..........................     1,014         (357)        291
          Accounts payable and accrued liabilities ..........     3,439        3,360         271
                                                               ----------  ----------  ----------
Net used in operating activities ............................   (18,645)     (10,256)    (10,696)

CASH FLOW FROM INVESTING ACTIVITIES
Purchases of short-term investments .........................  (151,582)     (87,728)    (41,618)
Sales/maturities of short-term investments ..................    78,348       68,562      50,006
Purchases of property and equipment, net ....................    (2,440)      (2,061)     (3,683)
                                                               ----------  ----------  ----------
Net cash flows (used in) provided by investing activities ...   (75,674)     (21,227)      4,705

CASH FLOW FROM FINANCING ACTIVITIES
Issuance of Common Stock ....................................    93,360       41,016         433
Proceeds received from long-term obligations ................     1,741          981       2,500
Principal payments on long-term obligations .................      (984)        (957)     (1,006)
Payments received on notes receivable from stockholders .....        15            -           1
                                                               ----------  ----------  ----------
Net cash flows provided by financing activities .............    94,132       41,040       1,928
                                                               ----------  ----------  ----------
Net (decrease) increase in cash and cash equivalents ........      (187)       9,557      (4,063)
Cash and cash equivalents at beginning of the period ........    21,265       11,708      15,771
                                                               ----------  ----------  ----------
Cash and cash equivalents at end of the period ..............  $ 21,078     $ 21,265    $ 11,708
                                                               ==========  ==========  ==========

SUPPLEMENTAL DISCLOSURES
Supplemental disclosures of cash flow information:
      Interest paid .........................................  $    228     $    231    $    150
      Taxes paid ............................................       302            -           1

Schedule of noncash investing and financing activities:
      Conversion of note receivable to investment in NPI ....         -            -    $  1,401
      Conversion of NPI Preferred Stock to investment in NPI          -            -       3,855




                             See accompanying notes.




                          NEUROCRINE BIOSCIENCES, INC.
                 NOTES TO THE CONSOLIDATED FINANCIAL STATEMENTS
                                December 31, 2000


NOTE 1.  ORGANIZATION AND SUMMARY OF SIGNIFICANT ACCOUNTING POLICIES

         Business  Activities.  Neurocrine  Biosciences,  Inc. (the "Company" or
"Neurocrine")  was  incorporated  in  California  on  January  17,  1992 and was
reincorporated in Delaware in March 1996.  Neurocrine is a leading  neuroscience
company  focused on the  discovery and  development  of novel  therapeutics  for
neuropsychiatric,   neuroinflammatory   and   neurodegenerative   diseases   and
disorders.  The Company's  neuroscience,  endocrine and  immunology  disciplines
provide a unique biological  understanding of the molecular  interaction between
central nervous, immune and endocrine systems for the development of therapeutic
interventions for anxiety, depression, insomnia, stroke, malignant brain tumors,
multiple sclerosis, obesity and diabetes.

         Principles of  Consolidation.  The  consolidated  financial  statements
include the accounts of the Company and its wholly owned  subsidiary,  Northwest
NeuroLogic,  Inc. ("NNL").  Significant  intercompany  accounts and transactions
have been eliminated in consolidation. In December 1999, NNL was merged with and
into the Company.

         Use of Estimates. The preparation of financial statements in conformity
with  generally  accepted  accounting  principles  requires  management  to make
estimates  and  assumptions  that affect the amounts  reported in the  financial
statements and the  accompanying  notes.  Actual results could differ from those
estimates.

         Cash Equivalents.  The Company considers all highly liquid  investments
with a maturity of three months or less when purchased, to be cash equivalents.

         Short-Term Investments Available-for-Sale. In accordance with Statement
of Financial  Accounting  Standards (SFAS) No. 115, "Accounting for Certain Debt
and   Equity   Securities,"    short-term    investments   are   classified   as
available-for-sale.  Available-for-sale  securities  are  carried at fair value,
with the  unrealized  gains and losses  reported in  comprehensive  income.  The
amortized cost of debt securities in this category is adjusted for  amortization
of premiums  and  accretion  of discounts  to  maturity.  Such  amortization  is
included in investment  income.  Realized gains and losses and declines in value
judged to be other-than-temporary,  if any, on available-for-sale securities are
included in investment  income or loss. The cost of securities  sold is based on
the  specific  identification  method.  Interest  and  dividends  on  securities
classified as available-for-sale are included in interest income.

         The Company invests its excess cash primarily in investment  grade debt
instruments,  marketable  debt  securities  of  U.S.  government  agencies,  and
high-grade commercial paper.  Management has established  guidelines relative to
diversification and maturities that maintain safety and liquidity.

         Property and  Equipment.  Property and  equipment  are carried at cost.
Depreciation  and  amortization  are provided over the estimated useful lives of
the assets, ranging from three to ten years, using the straight-line method.

         Licensed  Technology and Patent Application Costs.  Licensed technology
consists of  worldwide  licenses to patents  related to the  Company's  platform
technology,  which are capitalized at cost and amortized over periods of 7 to 11
years.  These costs are regularly  reviewed to determine that they include costs
for patent  applications the Company is pursuing.  Costs related to applications
that are not being actively  pursued are evaluated under  Accounting  Principles
Board (APB) Statement 17 "Intangible  Assets" and are adjusted to an appropriate
amortization  period,  which generally  results in immediate  write-off.  Assets
written-off   during  2000  had  a  net  book  value  of  $80,000.   Accumulated
amortization   at  December  31,  2000  and  1999  was  $753,000  and  $685,000,
respectively.

         Impairment  of  long-lived   assets.   In  accordance  with  SFAS  121,
"Accounting for the Impairment of Long-Lived Assets and for Long-Lived Assets to
be Disposed of," if indicators of  impairment  exist,  the Company  assesses the



recoverability  of the affected  long-lived  assets by  determining  whether the
carrying  value of such  assets can be  recovered  through  undiscounted  future
operating  cash flows.  If  impairment is  indicated,  the Company  measures the
amount of such  impairment by comparing  the carrying  value of the asset to the
present value of the expected  future cash flows  associated with the use of the
asset. While the Company's current and historical operating and cash flow losses
are indicators of impairment,  the Company  believes the future cash flows to be
received from the long-lived  assets will exceed the assets carrying value,  and
accordingly  the  Company  has not  recognized  any  impairment  losses  through
December 31, 2000.

         Industry Segment and Geographic Information.  The Company operates in a
single industry  segment - the discovery and development of therapeutics for the
treatment of neurologic and endocrine diseases and disorders. The Company has no
foreign operations.

         Research  and   Development   Revenue  and  Expenses.   Revenues  under
collaborative  research  agreements  and grants are recognized as research costs
are  incurred  over the period  specified  in the  related  agreement  or as the
services are performed.  These agreements are on a best-efforts basis and do not
require  scientific  achievement  as a  performance  obligation  and provide for
payment to be made when costs are incurred or the services  are  performed.  All
fees are nonrefundable to the collaborators.  Up-front,  nonrefundable  payments
for license fees and advance payments for sponsored  research  revenues received
in excess of amounts earned are classified as deferred revenue and recognized as
income over the period earned. Milestone payments are recognized as revenue upon
achievement of pre-defined  scientific  events.  Revenues from government grants
are recognized  based on a  percentage-of-completion  basis as the related costs
are  incurred.  The  Company  recognizes  revenue  only  on  payments  that  are
nonrefundable and when the work is performed. Research and development costs are
expensed as incurred.  Such costs include  proprietary  research and development
activities  and expenses  associated  with  collaborative  research  agreements.
Research and  development  expenses  relating to  collaborative  agreements  and
grants were approximately  $10.1 million,  $7.2 million and $12.0 million during
2000, 1999 and 1998, respectively.

         Stock-Based  Compensation.  As permitted by SFAS 123,  "Accounting  for
Stock-Based Compensation," the Company has elected to follow APB Opinion No. 25,
"Accounting  for Stock  Issued to  Employees",  and related  Interpretations  in
accounting for  stock-based  employee  compensation.  Deferred  compensation  is
recorded  for  employee  options only in the event that the fair market value of
the stock on the date of the option  grant  exceeds  the  exercise  price of the
options.  The deferred  compensation is amortized over the vesting period of the
options.

         In March 2000, the Financial  Accounting  Standards Board (FASB) issued
Interpretation No. 44, (FIN 44), "Accounting for Certain Transactions  Involving
Stock Compensation - An Interpretation of APB 25". This interpretation clarifies
the  definition  of employee  for  purposes of applying APB 25, the criteria for
determining whether a plan qualifies as a non-compensatory  plan, the accounting
consequence of various  modifications  to the terms of a previously  fixed stock
option or award, and the accounting for an exchange of stock compensation awards
in a business  combination.  FIN 44 was  effective  and the Company  adopted the
interpretation  on July 1, 2000. The adoption did not have a material  impact on
the Company's consolidated results of operations.

         Deferred  charges  for  options  granted  to  non-employees   has  been
determined in  accordance  with SFAS 123 and EITF 96-18 as the fair value of the
consideration  received  or the fair  value of the  equity  instruments  issued,
whichever is more reliably  measured.  Deferred  charges for options  granted to
non-employees are periodically remeasured as the underlying options vest and are
included in deferred compensation in the financial statements.

         Earnings Per Share. Basic and diluted earnings per share are calculated
in  accordance  with SFAS 128,  "Earnings  per  Share." All  earnings  per share
amounts  for all  periods  have  been  presented,  and where  appropriate,  were
restated to conform to the requirements of SFAS 128.

         Comprehensive Income.  Comprehensive income is calculated in accordance
with SFAS 130,  "Comprehensive Income." The Statement requires the disclosure of
all  components  of  comprehensive  income,  including net income and changes in
equity  during a period from  transactions  and other  events and  circumstances



generated from  non-owner  sources.  The Company's  other  comprehensive  income
consisted of gains and losses on short-term  investments  and is reported in the
consolidated statement of stockholders' equity.

         Reclassifications.  Certain  reclassifications  have been made to prior
year  amounts to conform to the  presentation  for the year ended  December  31,
2000.

         Impact of Recently Issued Accounting  Standards.  In December 1999, the
SEC issued Staff  Accounting  Bulletin  (SAB) No. 101,  "Revenue  Recognition in
Financial  Statements." SAB 101 provides guidance in applying generally accepted
accounting principles to revenue recognition in financial statements,  including
the recognition of  nonrefundable  up-front fees received in conjunction  with a
research and development  arrangement.  The adoption of this  pronouncement  was
required effective with the fourth quarter of 2000.

         As  required  by  the  adoption,  the  Company  reviewed  all  up-front
payments,  license fees and milestones  received in the current and prior years.
Up-front  payments have been received for program cost  reimbursements  incurred
during a negotiation  period.  License fees are received in exchange for a grant
to use our  proprietary  technologies  on an  as-is  basis,  for the term of the
collaborative  agreement.   Milestones  are  received  for  specific  scientific
achievements   determined   at  the  beginning  of  the   collaboration.   These
achievements are remote and  unpredictable at the onset of the collaboration and
are based on the success of scientific efforts.

         Based on that  review,  the  Company  determined  that $4.2  million of
license fees  received  during 2000 were subject to the adoption of SAB 101. All
other fees received relate to agreements under which the research portion of the
collaboration  has  been  completed  or  the  agreements  have  been  terminated
entirely.  In accordance  with APB 20, the adoption of SAB 101 was recognized by
including the cumulative effect of the change in accounting principle in the net
loss for the fourth  quarter of 2000.  The  otherwise  reported net loss for the
year ended December 31, 2000 was increased by approximately $3.8 million.  These
license fee revenues  were  deferred and will be amortized as income at $915,000
in 2001,  $835,000 in 2002,  $828,000 in 2003,  $818,000 in 2004 and $409,000 in
2005.

         In June 1998,  the FASB  issued  SFAS  133,"Accounting  for  Derivative
Instruments  and  Hedging  Activities."  The  Company  expects  to adopt the new
Statement  effective January 1, 2001. This statement requires the recognition of
all  derivative  instruments as either assets or liabilities in the statement of
financial  position and the measurement of those  instruments at fair value. The
Company does not expect the adoption of this statement to have a material impact
on its results of operations or financial position.

         In  September  2000,  the FASB  issued SFAS No.  140,  "Accounting  for
Transfers and Servicing of Financial Assets and Extinguishments of Liabilities."
SFAS 140 provides accounting and reporting standards for transfers and servicing
of financial  assets and  extinguishments  of  liabilities  and is effective for
transfers and servicing of financial assets and  extinguishments  of liabilities
occurring after March 31, 2001. The adoption of SFAS 140 is not expected to have
a material impact on the Company's financial statements.


Note 2.  Short-Term Investments

         The  following is a summary of  short-term  investments  classified  as
available-for-sale securities (in thousands):

                                               Gross       Gross      Estimated
                                Amortized   Unrealized   Unrealized      Fair
                                   Cost        Gains      Losses        Value
                                   ----        -----      ------        -----
December 31, 2000
US Government securities ...... $  2,000       $  -       $  (3)      $  1,997
Corporate debt securities .....  141,331        264           -        141,595
                                --------       ----       ------      --------
         Total securities ..... $143,331       $264       $  (3)      $143,592
                                ========       ====       ======      ========



December 31, 1999
US Government securities ...... $  1,997       $  -       $ (24)      $  1,973
Corporate debt securities .....   68,100          7        (247)        67,860
                                 -------       ----       ------      --------
         Total securities ..... $ 70,097       $  7       $ (271)     $ 69,833
                                 =======       ====       =======     ========


         Gross  realized  gains  and  losses  were not  material  for any of the
reported periods. The amortized cost and estimated fair value of debt securities
by contractual maturity at December 31, 2000 are shown below (in thousands).



                                                     Amortized     Estimated
                                                       Cost        Fair Value
                                                     ---------     ---------
   Due in one year or less .......................  $  48,000     $  48,000
   Due after one year through four years .........     95,331        95,592
                                                    ---------     ---------
                                                    $ 143,331     $ 143,592
                                                    =========     =========


NOTE 3.  PROPERTY AND EQUIPMENT

         Property  and  equipment  at December  31, 2000 and 1999 consist of the
following (in thousands):

                                                  2000              1999
                                               ---------        ---------
    Land ..................................   $    5,003       $    5,299
    Furniture and fixtures ................        2,051            1,982
    Equipment .............................       11,179            9,046
    Leasehold improvements ................        1,113              875
                                               ---------        ---------
                                                  19,346           17,202
    Less accumulated depreciation .........       (8,046)          (6,021)
                                               ---------        ---------
    Net property and equipment ............    $  11,300        $  11,181
                                               =========        =========

         Furniture and equipment under capital leases were $8.5 million and $6.7
million at December 31, 2000 and 1999, respectively. Accumulated depreciation of
furniture  and  equipment  under  capital  leases  totaled $5.0 million and $4.0
million at December 31, 2000 and 1999,  respectively.  The Company  entered into
$1.8 million of additional capital leases during 2000 and $981,000 during 1999.


NOTE 4.  ACCRUED LIABILITIES

         Accrued  liabilities  at  December  31,  2000 and 1999  consist  of the
following (in thousands):

                                                     2000            1999
                                                  ---------      ---------
    Accrued employee benefits ................   $   2,992       $  1,331
    Accrued professional fees ................       1,229            270
    Accrued offering expenses ................         277          1,222
    Accrued development costs ................       6,199          1,828
    Taxes payable ............................          26             27
    Other accrued liabilities ................         412            391
                                                  ---------      --------
                                                  $ 11,135       $  5,069
                                                   ========      ========



NOTE 5.  LONG-TERM DEBT

         During 1997, the Company partially  financed the purchase of land under
a 5 year note  payable for  approximately  $747,000,  which bears  interest at a
floating rate of prime plus one quarter percent (9.75% and 8.75% at December 31,
2000  and  1999,   respectively).   The  note  is  repayable  in  equal  monthly
installments beginning February 1998.

         At  December  31,  2000,  the  balance  of the note was  $311,000.  The
repayment  schedule for the note is $149,000 for each year 2001 through 2002 and
$13,000 in the year 2003.


NOTE 6.  COMMITMENTS AND CONTINGENCIES

         Capital Lease  Obligations.  The Company has financed certain equipment
under capital lease obligations,  which expire on various dates through the year
2005 and bear interest at rates between 6.0% and 9.6%. The lease commitments are
repayable in monthly installments.

         Operating  Leases.  In May 1997,  the Company  purchased  two  adjacent
parcels of land in San Diego for $5.0 million.  In August 1997, the Company sold
one parcel to Science Park Center LLC, a California  limited  liability  company
(the "LLC"), of which the Company owns a nominal minority interest,  in exchange
for a note  receivable  in the amount of $3.5  million  plus  interest of 8.25%.
However,  for accounting  purposes,  this transaction does not qualify as a sale
under SFAS No. 98 and  therefore,  the entire  amount of the note  receivable is
included in land. The amount  included in land at December 31, 2000 and 1999 was
$3.5 and $3.8  million,  respectively.  The  second  parcel of land will be held
until such time as additional facilities are required.

         During 1998,  the LLC  constructed  an expanded  laboratory  and office
complex,  which was  leased by the  Company  under a  15-year  operating  lease,
commencing  September  1998. The Company has the option to purchase the facility
at any time  during the term of the lease at a  predetermined  price.  The lease
contains  a 4% per year  escalation  in base  rent  fees,  effective  with  each
anniversary.  The  Company  subleases  a portion  of the space to two  unrelated
parties.  In August 2001,  the first  sublease  will revert to a  month-to-month
lease. The second sublease will expire in September 2001.

         Repayment  schedules  for the capital lease  obligations  and operating
lease commitments at December 31, 2000 are as follows (in thousands):

                                                     Capital         Operating
   Fiscal Year:                                      Leases           Leases
   ------------                                      ------           ------

   2001 ..........................................  $   1,662       $    2,525
   2002 ..........................................      1,073            2,626
   2003 ..........................................        672            2,731
   2004 ..........................................        480            2,841
   2005 ..........................................        112            2,954
   Thereafter ....................................          -           26,926
                                                     --------       ----------
     Total minimum payments ......................   $  3,999        $  40,603
                                                                     =========
   Less:  amounts representing interest ..........       (440)
                                                     ---------
   Future minimum payments .......................      3,559
   Less:  current portion ........................     (1,438)
                                                     ---------
   Future payments on capital lease obligations ..   $   2,121
                                                     =========




         Rent  expense was $2.5  million,  $2.7 million and $2.4 million for the
years ended December 31, 2000, 1999 and 1998, respectively.  Sublease income was
$1.2 million,  $1.2 million and $837,000 for the years ended  December 31, 2000,
1999, and 1998, respectively.

         Future  minimum  sublease  income to be received  under  non-cancelable
subleases at December 31, 2000 will be $297,000 for the year ending December 31,
2001.

         Licensing  and  Research  Agreements.  The  Company  has  entered  into
licensing agreements with various universities and research organizations, which
are  cancelable at the option of the Company with terms ranging from 30-180 days
written notice.  Under the terms of these  agreements,  the Company has received
licenses to  technology,  or technology  claimed,  in certain  patents or patent
applications.  The  Company is  required  to pay  royalties  on future  sales of
products  employing the  technology  or falling  under claims of a patent,  and,
certain  agreements  require minimum royalty payments.  Certain  agreements also
require  the  Company  to  make  payments  upon  the  achievement  of  specified
milestones.  Due to the uncertainty of the pharmaceutical  development  process,
the  Company  continually  reassesses  the value of the license  agreements  and
cancels them as research efforts are discontinued on these programs.


Note 7.  Stockholders' Equity

         Common Stock  Issuances.  From inception  through 1996, the Company has
issued  Common  Stock in various  private  and public  offerings,  as well as to
corporate collaborators,  at prices between $5.00 and $30.00 per share resulting
in aggregate net proceeds of approximately $201.9 million. This total includes a
December 2000 public  offering,  in which the Company sold 3.2 million shares of
its  Common  Stock  at $30 per  share.  The net  proceeds  generated  from  this
transaction were $90.4 million.

         Options.  The Company has  authorized  6.5 million shares of its Common
Stock for issuance upon  exercise of options or stock  purchase  rights  granted
under the 1992 Incentive  Stock Option Plan,  1996 Director  Option Plan and the
1997 NNL Stock Option Plan.  These plans  provide for the grant of stock options
and  stock  purchase  rights to  officers,  directors,  and  employees  of,  and
consultants  and advisors to, the Company.  Options under these plans have terms
of up to 10 years  from the date of grant  and may be  designated  as  incentive
stock options or nonstatutory stock options under the plans.

         A  summary  of  the  Company's  stock  option  activity,   and  related
information for the years ended December 31 follows:



                                      2000               1999                   1998
                             -------------------- ------------------- --------------------
                                        Weighted             Weighted             Weighted
                              Options    Average   Options    Average   Options    Average
                                (in     Exercise     (in     Exercise     (in     Exercise
                             thousands)   Price   thousands)   Price   thousands)   Price
                             -------------------- ------------------- --------------------
                                                                 
Outstanding at January 1, ...   3,158   $ 5.91       2,793   $6.02      2,653      $5.84
Granted .....................   1,136    29.66       1,142    6.03        677       6.26
Exercised ...................    (354)    6.56        (412)   4.79        (81)      3.64
Canceled ....................     (29)   11.69        (365)   6.52       (456)      5.76
                             -------------------- ------------------- --------------------
Outstanding at December 31, .   3,911   $12.75       3,158   $5.91      2,793      $6.02
                             ==================== =================== ====================




         A summary of options outstanding as of December 31, 2000 follows:

                            Options Outstanding             Options Exercisable
- -------------- ----------------------------------------- -----------------------
                              Weighted
                               Average
Range           Outstanding   Remaining    Weighted     Exercisable   Weighted
of Exercise        as of     Contractual    Average       As of       Average
Prices           12/31/00        Life    Exercise Price 12/31/00  Exercise Price
- -------------- ---------------------------------------- ------------------------
$0.02 to $4.19    510          3.6          $ 2.47         447         $2.47
 4.25 to 4.94     544          5.7            4.47         416          4.35
 5.00 to 6.50     599          7.9            5.52         275          5.57
 6.56 to 7.38     498          6.7            7.17         398          7.22
 7.75 to 10.25    598          6.4            8.47         476          8.43
11.19 to 34.44    589          9.3            22.70         58          19.35
34.50 to 43.44    573          9.4            36.39         70          35.01
               ---------------------------------------- ------------------------
                 3,911         7.1          $12.75        2,140        $6.96

         The weighted  average fair values of the options  granted  during 2000,
1999 and 1998 were $20.51, $3.75 and $5.59, respectively.

         Pro forma  information  regarding net income (loss) is required by SFAS
No. 123 and has been determined as if the Company had accounted for its employee
stock options under the fair value method of that Statement.  The fair value for
these  options was estimated at the date of grant using a  Black-Scholes  option
pricing model using the following  weighted-average  assumptions  for 2000, 1999
and 1998,  respectively:  risk-free  interest  rates of 5.0%,  6.4% and 5.5%;  a
dividend  yield of 0.0% (for all  years),  volatility  factors  of the  expected
market price of the  Company's  common stock of .81, .74 and .88; and a weighted
average expected life of the option of 5 years (for all years presented).

         For purposes of pro forma disclosures,  the estimated fair value of the
options granted is amortized to expense over the options'  vesting  period.  The
pro forma  effect on net  losses  for  2000,  1999 and 1998 is not  likely to be
representative of the effects on reported income or loss in future years because
these amounts  reflect less than full vesting for options  granted  during these
periods.  The Company's pro forma  information  for the years ended December 31,
2000, 1999, and 1998 follows (in thousands, except for per share data):

                                                    2000        1999     1998
                                                ---------- ---------- ----------
 Net income (loss) as reported ...............  $(28,808)  $(16,822)  $(19,955)
 Earnings (loss) per share (diluted) .........     (1.30)     (0.88)     (1.10)

 Pro forma net income (loss) .................   (31,057)   (18,303)   (20,758)
 Pro forma earnings (loss) per share (diluted)     (1.40)     (0.96)     (1.14)

         Employee Stock  Purchase Plan. The Company has reserved  425,000 shares
of Common Stock for issuance  under the 1996 Employee  Stock  Purchase  Plan, as
amended  on May 24,  2000 (the  "Purchase  Plan").  The  Purchase  Plan  permits
eligible  employees to purchase  Common Stock  through  payroll  deductions at a
purchase price equal to 85% of, the lesser of the fair market value per share of
Common  Stock on the  enrollment  date or on the date on which  the  shares  are
purchased.  As of December 31, 2000, 267,000 shares have been issued pursuant to
the Purchase Plan.




         Warrants.  The Company  has  outstanding  warrants to purchase  356,000
shares of Common Stock at an exercise price of $10.50 per share.  These warrants
generally  expire in 2007. At December 31, 2000, all  outstanding  warrants were
exercisable.

         The following  shares of Common Stock are reserved for future  issuance
at December 31, 2000 (in thousands):

  Stock option plans ......................................  4,142
  Employee stock purchase plan ............................    158
  Warrants ................................................    356
                                                            ------
  Total ...................................................  4,656
                                                            ======

         Of the shares available for future issuance under the Plan, 3.9 million
are outstanding grants and 231,000 remain available for future grant.


NOTE 8.  ACQUIRED IN-PROCESS RESEARCH AND DEVELOPMENT AND LICENSES

         Northwest NeuroLogic, Inc. In May 1998, the Company acquired the assets
and liabilities of NNL in exchange for shares of the Company's  Common Stock and
stock options valued at $4.2 million.  Since the acquisition,  the operations of
NNL have been included in the Company's  consolidated  statements of operations.
The acquisition was accounted for as a purchase and,  accordingly,  the purchase
price was allocated to the assets acquired and the liabilities  assumed based on
the estimated  fair market values.  Substantially  all of the purchase price was
allocated to the in-process research and development. The value allocated to the
technology  was  then  expensed   because  it  had  not  reached   technological
feasibility and had no future alternative uses. The Company performed scientific
due diligence related to the acquired  projects,  and because they were based on
narrow  scientific  hypotheses,  the Company  concluded that neither program had
alternative future uses.

         The nature and  efforts  required to develop  the  acquired  in-process
research and development into  commercially  viable products include research to
identify a clinical candidate,  preclinical  development,  clinical testing, FDA
approval and commercialization.  This process may cost in excess of $100 million
and can take as long as 10 years to complete.  It is also important to note that
if a clinical candidate is identified, the further development of that candidate
can be halted or abandoned at any time due to a number of factors. These factors
include,  but are not limited  to,  funding  constraints,  safety or a change in
market demand.

         Because of limited  financial  resources,  the  Company's  strategy  to
develop  some of its  programs is to enter into  collaborative  agreements  with
major pharmaceutical companies. Through these collaborations,  the Company could
partially  recover its research  costs through  contract  research and milestone
revenues.  The  collaborators  would  then be  financially  responsible  for all
clinical development and commercialization costs.

         In May 1998,  when the Company  acquired  the  in-process  research and
development  programs  from NNL, it  estimated  the costs to identify a clinical
candidate and provide minimal research  support during the clinical  development
stages for the melanocortin  receptor program to be $15.4 million over an 8-year
period.  Costs to identify a clinical  candidate  and provide  minimal  research
support  during the clinical  development  stages of the  excitatory  amino acid
transporters  program were estimated at $22.4 million.  Estimated  revenues from
the  collaborative  arrangements  were  anticipated  to reduce the Company's net
costs.  The  clinical  development  and  commercialization   costs  were  to  be
completely funded by the collaborator.

         During fiscal year 2001, the Company  anticipates  that its gross costs
for continued  research on these  programs will  approximate  $5.0 million.  The
Company  cannot be certain  that its  research  efforts  will result in clinical
candidates  for either  compound.  The  Company  intends to  collaborate  on the



melanocortin  receptor technology.  The Company would expect the collaborator to
then be responsible for the clinical development, commercialization and funding.
The  excitatory   amino  acid   transporters   program  is  currently   under  a
collaborative  agreement  with  Wyeth-Ayerst.  Consequently,  the Company cannot
estimate the time or resources  Wyeth-Ayerst  will commit to the  development of
this program.

         The following  are pro forma  unaudited  results of operations  for the
year ended  December  31,  1998 (in  thousands,  except per share  data) had the
purchase  of NNL  been  consummated  as of  January  1,  1998.  This  pro  forma
information is not necessarily  indicative of the actual results that would have
been achieved nor is it necessarily indicative of future results.

      Revenues ............................................   $16,325
      Net loss ............................................   (20,013)
      Loss per share basic and diluted ....................   $ (1.09)

         Other.  During 1998, the Company  purchased  licenses for  technologies
relating to insomnia and brain cancer in the amount of $710,000.  These projects
were  in the  early  stages  of  development,  have  not  reached  technological
feasibility and have no known alternative uses. Consequently, the costs of these
licenses were expensed.

         The insomnia and brain cancer compounds are both in the early stages of
clinical  testing.  During 2001, the Company expects to spend  approximately $35
million  on  additional  clinical  testing  of the  brain  cancer  and  insomnia
compounds.  The  Company  expects  the  clinical  testing of both  compounds  to
continue  for at least the next two  years,  but its  efforts  may not result in
commercially   viable  products.   If  the  Company's  efforts  were  completely
successful and it did not collaborate on these  compounds,  it is estimated that
each compound  could cost an  additional  $50 - $150 million and take up to five
years to reach commercial viability.


Note 9.  Collaborative Research and Development Agreements

         Taisho  Pharmaceuticals Co., Ltd. In December 1999, the Company entered
into an agreement with Taisho  Pharmaceutical  Co., Ltd.  (Taisho)  providing an
exclusive  option to obtain European,  Asian and North American  development and
commercialization rights for our altered peptide ligand product (APL), NBI-6024,
for Type 1 Diabetes.  In June 2000, Taisho exercised its option as to Europe and
Asia.  In  November  2000,  the  agreement  was  amended  to  include  worldwide
development  and  commercialization  rights.  Taisho  and the  Company  formed a
steering committee to oversee the worldwide development of NBI-6024.  Under this
agreement,  the Company is entitled to receive license fees, milestone payments,
and after the November  amendment,  sponsored research funding and reimbursement
of 100% of worldwide development expenses. In addition, the Company will receive
payments  on  product  sales in Europe  and  Japan  for the term of the  patents
covering  NBI-6024  subject to  adjustment  for payments to third  parties.  The
Company is also  entitled to receive up to $43.0  million for  milestones,  plus
additional  amounts for research funding and  reimbursement of development costs
and potential sublicense fees. As of December 31, 2000, the Company has received
$2.0 million for the exclusive  negotiating option, $3.0 million in license fees
for the European  and Asian  commercialization  rights,  $1.0 million in license
fees for the remaining  worldwide rights, $4.0 million in milestone payments and
$829,000 in reimbursement  of development  costs. The license fees were deferred
and are being  recognized as revenues over the life of the agreement at $319,000
in 2000, $818,000 in each of the years 2001 through 2004 and $409,000 in 2005.

         Wyeth-Ayerst Laboratories. In January 1999, the Company entered into an
agreement with  Wyeth-Ayerst  Laboratories  (Wyeth-Ayerst),  the  pharmaceutical
division of American Home Products Corporation, on the research, development and
commercialization of compounds which modulate excitatory amino acid transporters
(EAATs) for the treatment of neurodegenerative and psychiatric  diseases.  EAATs
are part of the family of  neurotransmitter  transporters and play a key role in
regulating the actions of neurotransmitters and brain function.

         The  agreement,  valued  at  up  to  $80.3  million  if  a  product  is
commercialized,   includes:   sharing  proprietary  technologies,   funding  for



research, payments for milestones reached, plus royalties on sales from products
resulting from the collaboration.  Under the terms of the agreement,  Neurocrine
expects to receive  three to five years of funding for research and  development
as well as worldwide  royalties on commercial sales of products that result from
the  collaboration.  Wyeth-Ayerst  will also provide  Neurocrine  with access to
chemical libraries for screening within the collaborative  field. As of December
31, 2000, the Company received $6.0 million in sponsored research payments, $3.0
million for the  achievement  of four  milestones  and $50,000 in license  fees,
which are being deferred and recognized over the life of the agreement.

         Eli Lilly and Company.  In October  1996,  the Company  entered into an
agreement with Eli Lilly and Company (Eli Lilly) under which Neurocrine received
three years of sponsored  research payments totaling $17.2 million.  The Company
is also entitled to milestone  payments for certain  development  and regulatory
accomplishments. The Company will have the option to receive co-promotion rights
and share profits from commercial  sales of select products that result from the
collaboration  in the U.S.  or receive  royalties  on U.S.  product  sales.  The
Company will receive royalties on product sales for the rest of the world.

         The  collaborative  research  portion of the agreement was completed as
scheduled in 1999. The Company will continue to receive  milestone  payments and
royalties upon the successful  continuation  of the  development  portion of the
agreement, if any.

         Janssen Pharmaceutica, N.V. In January 1995, the Company entered into a
research and development agreement with Janssen  Pharmaceutica,  N.V. (Janssen).
Under the  Janssen  agreement,  the  Company is  entitled to receive up to $10.0
million in milestone  payments for the  indications  of anxiety,  depression and
substance  abuse,  and up to $9.0 million in additional  milestone  payments for
other  indications.  The  Company  has granted  Janssen an  exclusive  worldwide
license to manufacture and market products. In exchange, the Company is entitled
to receive  royalties  on  worldwide  product  sales and has  certain  rights to
co-promote  such products in North America.  Janssen is responsible  for funding
all clinical development and marketing activities,  including  reimbursements to
Neurocrine  for its  promotional  efforts,  if any. As of December 31, 1998, the
Company has received  $2.0 million in up-front  license  fees,  $9.7 million for
three years of sponsored  research and $3.5 million in milestone  payments.  The
collaborative  research  portion of the  agreement was completed as scheduled in
1997 with the selection of a clinical candidate and the commencement of clinical
trials  in  Europe.  There  were no  additional  revenues  received  under  this
agreement in 1999 or 2000.

         In  September  1999,  the  Company  signed  an  amendment  to its  1995
agreement with Janssen to identify new  corticotropin-releasing  factor receptor
antagonist compounds. The amendment provides for a new sponsored research period
to begin  April 1999 and  conclude  in  February  2001.  All other  terms of the
agreement will be governed by the original  agreement  signed in 1995. Under the
amendment,  the Company will receive $5.0 million in sponsored research funding,
up to $3.5 million in milestone  achievements  and  reimbursement of all outside
and third party costs associated with the project.  As of December 31, 2000, the
Company  has  received  $4.6  million in  sponsored  research  and  $755,000  in
reimbursements of third party costs.

         In  April  2000,  Janssen  discontinued  development  of  the  compound
licensed under the 1995 agreement and replaced it with a back-up compound, which
resulted  from  research  under the 1999  amendment.  Since the new  compound is
subject to the terms of the original  agreement,  the Company  will  continue to
receive milestone payments and royalties upon the successful continuation of the
development  portion of the  agreement.  Janssen has the right to terminate  the
Agreement upon six months notice.  However,  in the event of termination,  other
than  termination  by  Janssen  for cause or as a result of the  acquisition  of
Neurocrine,  all product and technology rights become the exclusive  property of
Neurocrine

         Novartis.  In January 1996, the Company  entered into an agreement with
Novartis under which Novartis paid the Company $5.0 million in up-front  license
fees and was obligated to provide  Neurocrine  with $7.0 million in research and
development  funding during the first two years of the  agreement.  In addition,
the Company was eligible to receive up to $15.5 million in further  research and
development  funding  thereafter.  As of  December  31,  1999,  the  Company has



received $18.8 million in sponsored  research and development  payments and $9.1
million of milestone payments.

         On  July 7,  1999,  Novartis  exercised  its  right  to  terminate  the
agreement,  effective  January 7, 2000. As a result,  Neurocrine  reacquired the
worldwide rights to its multiple sclerosis compound, MSP771.


Note 10.  Related Party Transactions

         Neuroscience Pharma, Inc. In March 1996, the Company along with a group
of Canadian institutional  investors (the "Canadian Investors") established NPI.
The  Company's  contribution  was to license  certain  technology  and  Canadian
marketing rights to NPI. The Canadian Investors  contributed  approximately $9.5
million in cash in exchange for shares of NPI  Preferred  Stock (the  "Preferred
Shares"), which was convertible into shares of the Company's Common Stock at the
option of the Canadian Investors.  In addition,  the Canadian Investors received
warrants  exercisable  for 383,875  shares of the  Company's  Common Stock at an
exercise  price of $10.50 per share and may be  eligible  to receive  additional
warrants upon the attainment of certain additional  funding.  As of December 31,
2000, 29,625 warrants have been exercised.

         During 1997 and 1998, the Investors converted their Preferred Shares to
shares of the  Company's  Common  Stock.  As a result,  the Company  recorded an
investment  in NPI equal to the market  value of Common Stock issued in exchange
for the Preferred Shares and has recognized its  proportionate  share of the NPI
net losses in  accordance  with the equity method of  accounting.  Equity in NPI
losses totaled $764,000 and $3.4 million in 1999 and 1998, respectively.

         During 1996, the Company  entered into a sponsored  research  agreement
with NPI. The terms of the agreement called for NPI to fund additional  research
efforts on  technologies  licensed to NPI by the  Company.  Associated  with the
costs of research on those certain programs,  the Company recognized revenues of
$491,000 and $3.6 million during 1999 and 1998, respectively.

         The Preferred Shares were redeemable for cash at the Company's  option.
The  redemption  feature of the  Preferred  Shares  limited  their  value to the
balance  of cash and  cash  equivalents  maintained  by NPI.  Consequently,  the
Company reduced the value of its NPI investment by $647,000 during 1999 and $3.8
million  during 1998.  The balance of the Company's  investment in NPI was $0 at
December 31, 1999, and $1.4 million at December 31, 1998.

         In December  1999,  the Company sold its  investment in NPI in exchange
for cash,  receivables and potential royalties on worldwide sales resulting from
certain of NPI's future products. The Company recorded a gain of $526,000 on the
sale of this investment.  The gain was calculated using the total  consideration
of cash and receivables, less the carrying value of the NPI investment. No value
was  assigned  to  potential  royalties  on  future  product  sales  due  to the
uncertainty of this event. This  transaction,  as well as those discussed above,
is included in "Equity in NPI losses and other adjustments, net" reported on the
Consolidated Statement of Operations.


Note 11.  Earnings per Share

         The  following  data show the amounts  used in  computing  earnings per
share  and the  effect on income  and the  weighted-average  number of shares of
dilutive  potential  common  stock (in  thousands,  except for earning per share
data):



                                                     Year Ended December 31,
                                             -----------------------------------
                                                2000         1999         1998
                                             ---------    ---------    ---------
Numerator:
  Net income (loss) ......................   $(28,808)    $(16,822)    $(19,955)
  Effect of dilutive securities ..........          -          -            -
                                             ---------    ---------    ---------
  Numerator for earnings (loss) per share    $(28,808)    $(16,822)    $(19,955)
                                             =========    =========    =========
Denominator:
  Denominator for basic earnings
   (loss) per share ......................     22,124       19,072       18,141
  Effect of dilutive securities:
      Employee stock options .............         **           **           **
      Convertible preferred stock ........         **           **           **
      Warrants ...........................         **           **           **
                                             --------      --------    ---------
  Dilutive potential of common shares ....         **           **           **
                                             --------      --------    ---------
Demoninator for diluted earnings
  (loss) per share                             22,124       19,072       18,141
                                             ========      ========    =========

  Basic earnings (loss) per share ........   $  (1.30)    $  (0.88)    $  (1.10)
  Diluted earnings (loss) per share ......   $  (1.30)    $  (0.88)    $  (1.10)

                                            **Antidilutive

Note 12.  Income Taxes

         At December 31, 2000, the Company had Federal and California income tax
net  operating  loss  carry-forwards  of  approximately  $43.1 million and $24.3
million,  respectively.  The Federal and California tax loss carry-forwards will
begin to expire in 2010 and 2003, respectively,  unless previously utilized. The
Company also has Federal and California  research tax credit  carry-forwards  of
approximately $6.0 million and $2.9 million,  respectively,  which will begin to
expire in 2007 and 2012,  respectively,  unless previously utilized. The Company
has Federal  Alternative  Minimum  Tax credit  carry-forwards  of  approximately
$257,000, which will carry-forward indefinitely.

         Pursuant to Internal  Revenue Code Sections 382 and 383,  annual use of
the  Company's  net  operating  loss and  credit  carry-forwards  may be limited
because of  cumulative  changes in  ownership of more than 50%,  which  occurred
during 1992 and 1993.  However,  the Company  does not believe such changes will
have a material impact upon the utilization of these carry-forwards.

         Significant  components  of the  Company's  deferred  tax  assets as of
December  31,  2000 and 1999 are shown  below.  A valuation  allowance  of $27.2
million and $13.5 million at December 31, 2000 and 1999, respectively, have been
recognized to offset the net deferred tax assets as  realization  of such assets
is  uncertain.  Amounts  are  shown  in  thousands  as of  December  31,  of the
respective years:

                                                   2000           1999
                                                ---------      ----------
 Deferred tax assets:
   Net operating loss carry-forwards .........  $  16,487       $  7,400
   Tax credit carry-forwards .................      8,140          4,649
   Capitalized research and development ......      2,098            935
   Other, net ................................        479            520
                                                ---------      ---------
 Total deferred tax assets ...................     27,204         13,504
 Valuation allowance .........................    (27,204)       (13,504)
                                                ----------     ----------
 Net deferred tax assets .....................  $       -      $       -
                                                ==========     ==========


         The  provision  for income  taxes on earnings  subject to income  taxes
differs from the statutory federal rate at December 31, 2000, 1999 and 1998, due
to the following:



                                                   2000       1999       1998
                                                 --------   --------   --------
  Federal income taxes at 34% .................. $(9,692)   $(5,719)   $(6,785)
  State income tax, net of federal benefit .....       -          -          1
  Tax effect on non-deductible expenses ........     335        932      4,213
  Increase in valuation allowance ..............   9,357      4,787      2,572
                                                 -------    -------    -------
                                                 $     -    $     -    $     1
                                                 =======    =======    =======




NOTE 13.  SELECTED QUARTERLY FINANCIAL DATA (UNAUDITED)

         The following is a summary of the quarterly  results of operations  for
the years ended December 31, 2000 and 1999 (unaudited, in thousands,  except for
earnings per share data):





                                    ------------------------------------------------------------ -------------
                                                           Quarter Ended
                                    ------------------------------------------------------------   Year Ended
                                       Mar 31      Jun 30      Sep 30       Sep 30       Dec 31      Dec 31
                                                             As Reported  Restated (1)
                                    ------------------------------------------------------------ -------------
                                                                                  
Fiscal Year End 2000
Revenues ........................... $  2,778     $  2,942    $ 5,323      $  2,426     $ 6,442     $  14,588
Operating expenses .................   10,004       10,322     15,008        15,008      14,855        50,189
Net Loss ...........................   (6,047)      (5,192)    (8,135)      (11,032)     (6,537)      (28,808)

Earnings per share - Diluted ....... $ (0.28)     $  (0.24)   $ (0.37)     $ (0.50)     $ (0.29)    $   (1.30)
Shares used in the calculation of
  earnings per share - Diluted .....   21,771       21,897      22,032       22,032      22,789        22,124





                                    ----------------------------------------------------------------
                                                     Quarters Ended
                                    --------------------------------------------------  Year Ended
                                       Mar 31       Jun 30       Sep 30      Dec 31       Dec 31
                                    ----------------------------------------------------------------
                                                                        
Fiscal Year End 1999
Revenues ...........................  $  3,551    $   3,810    $   5,231   $  4,199    $ 16,791
Operating expenses .................     8,077        9,190       10,213      9,165      36,645
Net Loss ...........................    (4,089)      (4,637)      (4,407)    (3,689)    (16,822)

Earnings per share - Diluted .......  $  (0.22)   $   (0.24)   $  (0.23)   $  (0.19)   $  (0.88)
Shares used in the calculation of
  earnings per share - Diluted .....    18,955       18,961       19,006     19,361      19,072



    (1) During the fourth quarter of 2000, the Company adopted SAB 101, Revenue
     Recognition in Financial Statements.  SAB 101 provides, among other revenue
     items, guidance in the recognition of nonrefundable, up-front fees received
     in conjunction with a research and development  arrangement.  The result of
     the adoption of SAB 101 was to reduce  recognition  of license fee revenues
     reported  during the third quarter of 2000 by $2.9 million.  These revenues
     were deferred and will be recognized as income,  ratably over the estimated
     lives of the respective agreements. The adoption of SAB 101 did not require
     an adjustment for revenues recorded prior to December 31, 1999.