You should read the following discussion and analysis of our financial condition
and results of operations together with our unaudited financial information and
related notes included in this Form 10-Q and our consolidated financial
statements and related notes and other financial information in our Annual
Report on Form 10-K for the year ended December 31, 2019, which was filed with
the SEC on February 27, 2020 (the "2019 Form 10-K"). Some of the information
contained in this discussion and analysis or set forth elsewhere in this Form
10-Q, including information with respect to our plans and strategy for our
business, includes forward-looking statements that involve risks and
uncertainties. As a result of many factors, including those factors set forth in
Part II, Item 1A - Risk Factors in this Form 10-Q, our actual results could
differ materially from the results described in or implied by the
forward-looking statements contained in the following discussion and analysis.
                                       43

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

Table of Contents

Overview



We are a biotechnology company pioneering messenger RNA (mRNA) therapeutics and
vaccines to create a new generation of transformative medicines to improve the
lives of patients. mRNA medicines are designed to direct the body's cells to
produce intracellular, membrane, or secreted proteins that have a therapeutic or
preventive benefit with the potential to address a broad spectrum of diseases.
Our platform builds on continuous advances in basic and applied mRNA science,
delivery technology, and manufacturing, providing us the capability to pursue in
parallel a robust pipeline of new development candidates. We are developing
therapeutics and vaccines for infectious diseases, immuno-oncology, rare
diseases, autoimmune diseases and cardiovascular diseases, independently and
with our strategic collaborators.

Within our platform, we develop technologies that enable the development of mRNA
medicines for diverse applications. When we identify technologies that we
believe could enable a new group of potential mRNA medicines with shared product
features, we call that group a "modality." While the programs within a modality
may target diverse diseases, they share similar mRNA technologies, delivery
technologies, and manufacturing processes to achieve shared product features.
The programs within a modality will also generally share similar pharmacology
profiles, including the desired dose response, the expected dosing regimen, the
target tissue for protein expression, safety and tolerability goals, and
pharmaceutical properties. Programs within a modality often have correlated
technology risk, but because they pursue diverse diseases they often have
uncorrelated biology risk. We have created six modalities to date:

•prophylactic vaccines;
•cancer vaccines;
•intratumoral immuno-oncology;
•localized regenerative therapeutics;
•systemic secreted and cell surface therapeutics; and
•systemic intracellular therapeutics.

In 2019, we designated our prophylactic vaccines and systemic secreted and cell
surface therapeutics modalities as our "core modalities" based on positive Phase
1 data from our infectious disease vaccine portfolio, including our
cytomegalovirus, or CMV, vaccine and chikungunya antibody program. In these core
modalities, our strategy is to invest in additional development candidates using
our accumulated innovations in technology, our process insights and our
preclinical and clinical experience. As such, we have brought five new
development candidates forward in early 2020: a SARS-CoV-2 vaccine,
interleukin-2, or IL-2, programmed death-ligand 1, or PD-L1, a pediatric
Respiratory Syncytial Virus, or RSV vaccine, and an Epstein-Barr Virus, or EBV
vaccine, as part of our mission to use our technology to advance global public
health. Our exploratory modalities continue to be a critical part of advancing
our strategy to maximize the application of our potential mRNA medicines.

In response to the global coronavirus pandemic, we are pursuing the rapid
development and manufacture of our vaccine candidate, mRNA-1273, for the
treatment of SARS-CoV-2, the novel strain of coronavirus that causes COVID-19,
in collaboration with the Vaccine Research Center and Division of Microbiology
and Infectious Diseases of the National Institute of Allergy and Infectious
Diseases ("NIAID"), part of the National Institutes of Health ("NIH"). The
progress of mRNA-1273 during 2020 has resulted in the need for us to devote
significant resources toward the development and manufacture of this product.
Significant capital investment is necessary to prepare for the clinical
development, manufacturing and distribution of a vaccine at a scale necessary to
meet demand in a global pandemic environment. BARDA has committed to fund up to
$954.9 million to accelerate the clinical development and manufacturing process
scale-up of mRNA-1273. Under the terms of the agreement, BARDA will fund the
advancement of mRNA-1273 to FDA licensure and the scale-up of manufacturing
processes. The agreement does not contemplate any product stockpiling.

In May 2020, we completed a public offering of 17,600,000 shares of common stock
resulting in net proceeds of from the offering were $1.30 billion, net of
underwriting discount, commission and offering expenses. This additional funding
has enabled us to substantially expand our manufacturing network, purchase the
required capital equipment, hire appropriate global staff and secure the raw
materials and other consumables to manufacture substantial doses of mRNA-1273.

mRNA-1273 is currently being tested in several clinical trials in collaboration
with NIAID. We are in discussions with the United States government and many
other governmental agencies outside the United States related to the potential
sale of doses of mRNA-1273 should the product be approved by the relevant
regulatory requirements in each such country. As part of those discussions and
in certain cases, we may receive upfront deposits for our future mRNA-1273
vaccine supply, initially recorded as deferred revenue. During the three months
ended June 30, 2020, we recognized approximately $75.0 million in deferred
revenue in connection with such deposits. We will recognize revenue when revenue
recognition criteria have been met. As such, in the event that
                                       44
--------------------------------------------------------------------------------
  Table of Contents
mRNA-1273 is approved for distribution, we may expect to capitalize inventory
costs and record revenue related to product sales during 2020. Pre-launch
inventory costs are expensed in the period incurred and included in research and
development expense. Our initial product gross margin may be higher as our
pre-launch inventory costs will not be included in cost of goods sold.

COVID-19 has resulted in a significant burden of disease for the worldwide
population, especially those with pre-existing diseases and other comorbid
conditions such as cardiovascular disease, diabetes, chronic kidney disease,
chronic lung disease and obesity. In determining the pricing for a potentially
approved vaccine, we considered a health economic assessment framework that uses
standard metrics like the incremental cost effectiveness ratio (ICER) and the
standard willingness to pay thresholds as judged by quality adjusted life years
(QALY) gained from a therapy. This analysis does not reflect the costs of
factors like social disruption and economic loss. This assessment has resulted
in a potential assigned value to an effective COVID-19 vaccine on an ICER basis
with a QALY of $50,000 that ranges from $300 per 2-dose course to $725 per
2-dose course, with the value dependent on the age category and the epidemiology
of the disease, depending on whether the spread continues on the current
trajectory or there is increased transmission of COVID-19. With these values in
mind, our approach during the pandemic period has resulted in our working to
develop a safe and effective vaccine and to price that vaccine well below its
value during the pandemic period. To date, we have entered into smaller volume
agreements, primarily with governments, executed at $32-$37 per dose or $64-$74
per 2-dose course. It is expected that future larger volume agreements, if any,
may result in a lower price per dose. As and if the pandemic recedes and the
world enters an endemic period where a vaccine against COVID-19 is still
required, we expect that our vaccine will be priced in-line with other
innovative vaccines and will be dependent on market forces, including vaccine
efficacy and number of competitors. During the endemic period, we expect to use
traditional approaches to vaccine pricing, sale and distribution.

We have a diverse development pipeline, and the broad potential applications of
mRNA medicines have led us to raise significant capital and adopt a long-term
approach to capital allocation that balances near-term risks and long-term value
creation. As of June 30, 2020, we had cash, cash equivalents, and investments of
approximately $3.07 billion. We use this capital to fund operations and
investing activities for technology creation, drug discovery and clinical
development programs, infrastructure and capabilities to enable our research
engine and early development engine (which includes our Moderna Technology
Center), our digital infrastructure, creation of our portfolio of intellectual
property and administrative support.

Since our inception, we have incurred significant operating losses. Our net loss
was $514.0 million and $384.7 million for the years ended December 31, 2019 and
2018, respectively. Our net loss was $116.7 million and $240.9 million for the
three and six months ended June 30, 2020, respectively. As of June 30, 2020, our
accumulated deficit was $1.74 billion.

For the foreseeable future, we may continue to incur significant expenses and
operating losses in connection with our ongoing activities, including as we:
•continue our platform research and drug discovery and development efforts;
•build up our commercial operations and organization;
•conduct clinical trials for our investigational medicines;
•manufacture clinical trial materials and develop large-scale manufacturing
capabilities;
•seek regulatory approval for our investigational medicines;
•maintain, expand, and protect our intellectual property;
•hire additional personnel to support our program development effort to obtain
regulatory approval and secure additional facilities for operations; and
•continue to operate as a public company.
We do not expect to recognize revenue from the sale of potential mRNA medicines
unless and until we successfully complete clinical development and obtain
regulatory approval for one or more of our investigational medicines. If we seek
to obtain regulatory approval for and commercialize any of our investigational
medicines, we expect to incur significant commercialization expenses, which
include establishing a sales, marketing, manufacturing, and distribution
infrastructure globally.

As a result, we expect we will need substantial additional funding to support
our continued operations and pursue our growth strategy in addition to
commercial revenue that we may receive upon any sale of any of our products.
Until we can generate significant revenue from sales of our medicines, if ever,
we expect to finance our operations through a combination of public or private
equity offerings, structured financings and debt financings, government funding
arrangements, strategic alliances and marketing, manufacturing, distribution,
and licensing arrangements. We may be unable to raise additional funds or enter
into such other agreements on favorable terms, or at all. If we fail to raise
capital or enter into such agreements as, and when, needed, we may have to
significantly delay, scale back, or discontinue the development and
commercialization of one or more of our programs. Because of the numerous risks
and
                                       45
--------------------------------------------------------------------------------
  Table of Contents
uncertainties associated with pharmaceutical development, we are unable to
predict the timing or amount of expenses or when or if we will be able to
achieve or maintain profitability. Even if we are able to generate revenues from
the sale of our medicines, we may not become profitable. If we fail to become
profitable or are unable to sustain profitability on a continuing basis, then we
may be unable to continue our operations at planned levels and be forced to
reduce our operations.

In response to the global outbreak of coronavirus, we are pursuing the rapid
clinical testing and manufacture of our vaccine candidate, mRNA-1273. In May and
July 2020, we announced positive interim data from the NIH-led Phase 1 study. of
mRNA-1273. The Phase 2 placebo-controlled, dose-confirmation study of mRNA-1273
completed enrollment in early July 2020, and enrollment in the Phase 3 study of
mRNA-1273 began on July 27, 2020. We continue to commit financial resources and
personnel to the development of mRNA-1273, which may cause delays in or
otherwise negatively impact our other development programs.

The ultimate impacts of COVID-19 on our business are currently unknown. In March
2020, we announced that, based on the special concerns for the safety and health
of pediatric patients and their caregivers, and the risks of disruption to the
integrity of trials from COVID-19, we decided to pause new enrollment of our
Phase 1 rare disease clinical trials (mRNA-3704 for MMA, mRNA-3927 for PA) and
our age de-escalation trial for our pediatric respiratory vaccine (mRNA-1653 for
hMPV/PIV3). These decisions will be re-evaluated on an ongoing basis as the
COVID-19 situation evolves. We will continue to actively monitor the situation
and may take further precautionary and preemptive actions as may be required by
federal, state or local authorities or that we determine are in the best
interests of public health and safety and that of our patient community,
employees, partners, suppliers and stockholders. We cannot predict the effects
that such actions, or the impact of COVID-19 on global business operations and
economic conditions, may have on our business or strategy, including the effects
on our ongoing and planned clinical development activities and prospects, or on
our financial and operating results.

Our Pipeline
This section describes the pipeline that has emerged thus far from the
combination of our strategy, our platform, our infrastructure, and the resources
we have amassed.
Since we nominated our first program in late 2014, we and our strategic
collaborators have advanced in parallel a diverse development pipeline which
currently consists of 23 development candidates across our 22 programs. Since
December 2015 we have dosed approximately 2,000 subjects in our clinical trials
and in our Phase 3 trial of mRNA-1273 started in late July we expect to dose
30,000 people with our vaccine or placebo. Our diverse pipeline comprises
programs across six modalities and a broad range of therapeutic areas. A
modality is a group of potential mRNA medicines with shared product features,
and the associated combination of mRNA technologies, delivery technologies, and
manufacturing processes. Aspects of our pipeline have been supported through
strategic alliances, including with AstraZeneca plc, or AstraZeneca, Merck & Co,
Inc., or Merck, and Vertex Pharmaceuticals Inc., or Vertex, and
government-sponsored organizations and private foundations focused on global
health initiatives, the U.S. Biomedical Advanced Research and Development
Authority, or BARDA, the Defense Advanced Research Projects Agency, or DARPA,
the NIH, CEPI and the Bill & Melinda Gates Foundation, or the Gates Foundation.

The following chart shows our current pipeline of 23 development candidates
across our 22 programs, grouped into modalities-first the two core modalities
where we believe we have reduced the technology risk, followed by the four
exploratory modalities in which we are continuing to investigate the clinical
use of mRNA medicines.

                                       46

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

Table of Contents


                    [[Image Removed: mrna-20200630_g2.jpg]]

Abbreviations: IL-12, interleukin 12; IL-23, interleukin 23; IL-36?, interleukin
36 gamma; VEGF-A, vascular endothelial growth factor A.
The breadth of biology addressable using mRNA technology is reflected in our
current development pipeline of 22 programs. These span 26 different proteins or
protein complexes: 11 different antigens (including virus-like particles) for
infectious disease vaccines; two different cancer vaccines, one personalized
cancer vaccine addressing neoantigens and one for a shared cancer antigen; four
different immuno-modulator targets (including membrane and systemically secreted
proteins) for immuno-oncology programs; one secreted, local regenerative factor
for a heart failure program; four secreted or cell surface proteins of diverse
biology (an antibody, an engineered protein hormone, a secreted cytokine and a
cell surface receptor); and four intracellular enzymes for rare disease
programs.
                                       47

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

Table of Contents The diversity of proteins made from mRNA within our development pipeline is shown in the figure below. [[Image Removed: mrna-20200630_g3.jpg]] We have developed six modalities, which are summarized as follows:



•Prophylactic vaccines: Our prophylactic vaccines modality currently includes
eight programs, six of which have entered into clinical trials. Of these
programs, we have demonstrated desired pharmacology, in the form of
immunogenicity, in the positive Phase 1 clinical trials for the following eight
programs: H10N8 vaccine (mRNA-1440), H7N9 vaccine (mRNA-1851), RSV vaccine
(mRNA-1777), Chikungunya vaccine (mRNA-1388), human metapneumovirus (hMPV)/
parainfluenza virus type 3 (PIV3) vaccine (mRNA-1653), Zika vaccine (mRNA-1893),
CMV vaccine (mRNA-1647) and SARC-CoV-2 (mRNA-1273). We have an ongoing Phase 1
trial for the next generation Zika vaccine (mRNA-1893) and Merck is conducting a
Phase 1 trial for an additional RSV vaccine (mRNA-1172). Our SARS-CoV-2 vaccine
(mRNA-1273) is described in detail below. In addition to the eight programs
being developed, the H10N8 vaccine (mRNA-1440) and Chikungunya vaccine
(mRNA-1388) are two public health programs that are not being further developed
without government or other funding.

•Systemic secreted therapeutics: We have four systemic secreted and cell surface
therapeutics development candidates in our pipeline. Our secreted programs
include our antibody against Chikungunya virus (mRNA-1944), Relaxin (AZD7970)
for the treatment of heart failure, and IL-2 (mRNA-6231) for autoimmune
disorders. Our antibody against Chikungunya virus (mRNA-1944) has had positive
Phase 1 readouts to date and is currently being evaluated in an ongoing Phase 1
dose escalation study in healthy adults that is randomized and
placebo-controlled. The Phase 1 study evaluating escalating doses of mRNA-1944
administered via intravenous infusion in healthy adults has restarted after
COVID-19 disruptions. Both cohorts, one cohort at the 0.6 mg/kg dose with
steroid premedication and one cohort with two doses of 0.3 mg/kg (without
steroid premedication) given one week apart, are fully enrolled and all
participants have been dosed. The remaining programs for Relaxin (AZD7970) and
IL-2 (mRNA-6231) are currently in preclinical development. We have a cell
surface therapeutic program in this modality, PDL-1 (mRNA-6981) for autoimmune
hepatitis, which is currently in preclinical development.

•Cancer vaccines: We are currently developing two programs within our cancer
vaccines modality. Our personalized cancer vaccine program mRNA-4157 is being
developed in collaboration with Merck and is in a multiple-arm Phase 1 trial and
a randomized Phase 2 trial. A second personalized cancer vaccine, NCI-4650 was
being developed in collaboration with the National Cancer Institute, or NCI, and
was in an investigator-initiated single-arm Phase 1 trial which has been
completed. The two vaccines mRNA-4157 and NCI-4650 differ in the neoantigen
selection protocols used, but are otherwise
                                       48
--------------------------------------------------------------------------------
  Table of Contents
substantially the same. Our second program within this modality, mRNA-5671, is a
KRAS vaccine. Our strategic collaborator Merck has a Phase 1 clinical trial
ongoing for mRNA-5671.

•Intratumoral immuno-oncology: We have three programs in this modality. The
first program in this modality, OX40L (mRNA-2416), was designed to overcome
technological challenges in advancing this modality, including engineering the
mRNA sequence to minimize off-target effects, utilizing our proprietary lipid
nanoparticles (LNPs) to enhance safety and tolerability, and to demonstrate
expression of a membrane protein in patients. OX40L (mRNA-2416) is currently
being evaluated in an ongoing Phase 1/2 trial in the United States, and protein
expression has been demonstrated in a number of patients. Data from the
monotherapy arm of this ongoing study of mRNA-2416 showed that mRNA-2416 was
well-tolerated at all dose levels studied with the majority of adverse events
reported as grade 1 and 2 and no grade 3 adverse events reported. This data
supports the evaluation of intratumoral mRNA-2416 with the anti-PD-L1 inhibitor
durvalumab in solid tumors, which is ongoing in Part B of this study with a
focus on advanced ovarian carcinoma. Our second program, OX40L/IL-23/IL-36?
(Triplet) (mRNA-2752), has dosed patients in a Phase 1 study for the treatment
of advanced or metastatic solid tumor malignancies or lymphoma. Our third
program, IL-12 (MEDI1191), is being developed in collaboration with AstraZeneca.

•Localized regenerative therapeutics: Our localized VEGF-A program, AZD8601,
which is being developed by AstraZeneca, has completed a Phase 1a/b trial to
describe its safety, tolerability, protein production, and activity in diabetic
patients. The study has met its primary objectives of describing safety and
tolerability and secondary objectives of demonstrating protein production and
changes in blood flow post AZD8601 administration. In this trial, AZD8601 was
administered by intradermal injection in the forearm skin of patients for single
ascending doses. These data are consistent with studies previously conducted in
preclinical models. We believe these data provide clinical proof of mechanism
for our mRNA technology outside of the vaccine setting. AstraZeneca has
initiated a Phase 2a study of AZD8601 for VEGF-A for ischemic heart disease in
patients undergoing coronary artery bypass grafting (CABG) surgery with
moderately impaired systolic function, and the trial is ongoing.

•Systemic intracellular therapeutics: We have four systemic intracellular
therapeutics development candidates in our pipeline. Our intracellular programs
address methylmalonic acidemia, or MMA (mRNA-3704), propionic acidemia, or PA
(mRNA-3927), phenylketonuria, or PKU (mRNA-3283), and glycogen storage disorder
type 1a, or GSD1a (mRNA-3745). We have an open IND for mRNA-3704 for a planned
Phase 1/2 trial, and the FDA has also designated the investigation of mRNA-3704
for the treatment of isolated MMA due to MUT deficiency as a Fast Track
development program. We have an open IND for mRNA-3927 for a planned Phase 1/2
trial and this program has also been designated as a Fast Track development
program. PKU (mRNA-3283) is currently in preclinical development.

Our Vaccine Candidate Against SARS-CoV-2 (mRNA-1273)

In response to the global coronavirus pandemic, we are pursuing the rapid development and manufacture of our vaccine candidate, mRNA-1273, for the treatment of SARS-CoV-2, the novel strain of coronavirus that causes COVID-19, in collaboration with NIAID.

Preclinical Studies



On July 29, 2020, we announced the publication in The New England Journal of
Medicine of data from a preclinical study of mRNA-1273 in non-human primates. In
the study, immunogenicity and protective efficacy were assessed after a two-dose
vaccination schedule of 10 or 100 µg doses of mRNA-1273 or control given four
weeks apart (n=24; 8 per group). Four weeks after the second vaccination,
animals were challenged with high doses of SARS-CoV-2 through intranasal and
intratracheal routes.

After two vaccinations, the immune response observed in this non-human primate
study was consistent with the Phase 1 human study of mRNA-1273, also published
in The New England Journal of Medicine. At the 10 µg dose, the geometric mean
titer (GMT, ID50) measured in a pseudovirus (PsV) neutralization assay was 103,
similar to the GMT for a panel of convalescent sera reported previously (109),
and below the GMT achieved by mRNA-1273 in the Phase 1 human study at the 100 µg
dose (231) in the same PsV assay. At the higher dose in the non-human primates
(100 µg), neutralizing antibody titers increased further, with PsV GMT reaching
1,862. Vaccination also led to a significant increase in T cell responses,
primarily Th1 CD4 T cells.

Two doses of mRNA-1273 provided protection against lung inflammation following
viral challenge with SARS-CoV-2 in non-human primates at both the 10 µg and 100
µg dose levels. In addition, both the 10 µg and 100 µg dose groups demonstrated
protection against viral replication in the lungs, with the 100 µg dose also
protecting against viral replication in the nose of the animals. Of note, none
of the eight animals in the 100 µg group showed detectable viral replication in
the nose compared to six out of eight in the placebo group on day 2.

                                       49
--------------------------------------------------------------------------------
  Table of Contents
Preclinical results from a viral challenge study in mice conducted in
collaboration with NIAID and its academic partners are also available. In this
study, vaccination with mRNA-1273 prevented viral replication in the lungs of
mice challenged with SARS-CoV-2. Neutralizing titers in Phase 1 clinical trial
participants at the 25 µg and 100 µg dose levels (described below) were
consistent with neutralizing titers that were protective in the mouse and NHP
challenge models.

Phase 1 Study

A Phase 1 open-label study of mRNA-1273 is being conducted by the National
Institutes of Health (NIH). This study, which began on March 16, 2020,
originally enrolled 45 healthy adult volunteers ages 18 to 55 years and is
evaluating three dose cohorts (25 µg, 100 µg and 250 µg). An additional seven
cohorts in the Phase 1 study have since completed enrollment: a 50 µg cohort in
adults 18-55 (n=15), three cohorts of older adults (n=30, ages 56-70, 25 µg, 50
µg, and 100 µg) and three cohorts of elderly adults (n=30, ages 71 and above, 25
µg, 50 µg, and 100 µg).

On July 14, 2020, we announced the publication in The New England Journal of Medicine of an interim analysis of data from the original cohorts obtained through Day 57 in the Phase 1 study.



This interim analysis demonstrated that mRNA-1273 induced binding antibodies to
the full-length SARS-CoV-2 Spike protein (S) in all participants after the first
vaccination, with all participants seroconverting by Day 15. Dose dependent
increases in binding titers were seen across the three dose levels, and between
prime and boost vaccinations within the dose cohorts. After two vaccinations, at
Day 57, geometric mean titers exceeded those seen in convalescent sera obtained
from 38 individuals with confirmed COVID-19 diagnosis. Of the 38 individuals in
the convalescent sera group, 15% were classified as having severe symptoms
(hospitalization requiring intensive care and/or ventilation), 22% had moderate
symptoms and 63% had mild symptoms. Convalescent sera samples were tested using
the same assays as the study samples.

Neutralizing activity was assessed in two different assays, a live SARS-CoV-2
plaque-reduction neutralization test (PRNT) and a pseudovirus neutralization
assay (pseudotyped lentivirus reporter single-round-of-infection neutralization
assay, PsVNA). No participants had detectable live SARS-CoV-2 virus
neutralization or PsVNA responses prior to vaccination. After two vaccinations,
mRNA-1273 elicited robust neutralizing antibody titers. At Day 43, neutralizing
activity against SARS-CoV-2 (PRNT80) was seen in all evaluated participants. At
the Phase 3 selected dose of 100 µg, the geometric mean titer levels were
4.1-fold above those seen in reference convalescent sera (n=3). After the second
vaccination, PsVNA neutralizing antibody titers were detected in all
participants in all dose cohorts. The Day 57 geometric mean titers at the 100 µg
dose were 2.1-fold higher than those seen in convalescent sera (n=38)3. Strong
correlations were observed between the binding and neutralization assays, and
between the live virus and pseudovirus neutralization assays. A clear dose
response was seen in geometric mean titers between the 25 µg and 100 µg dose
levels, with minimal additional increases at the 250 µg dose. T-cell responses
were also evaluated at the 25 µg and 100 µg dose levels. Following second
vaccination, mRNA-1273 elicited Th1-biased CD4 T-cell responses without
significant elevation of Th2-biased CD4 T-cell responses.

mRNA-1273 was generally safe and well-tolerated, with no serious adverse events
reported through Day 57. Adverse events were generally transient and mild to
moderate in severity. The most notable adverse events were seen at the 250 µg
dose level, with three of those 14 participants (21%) reporting one or more
severe events. Solicited systemic adverse events were more common after the
second vaccination and occurred in seven of 13 (54%) participants in the 25 µg
group, all 15 participants in the 100 µg group and all 14 participants in the
250 µg group. The most commonly reported systemic adverse events following
second vaccination at the 100 µg dose were fatigue (80%), chills (80%), headache
(60%) and myalgia (53%), all of which were transient and mild or moderate in
severity. The most common solicited local adverse event at the 100 µg dose was
pain at the injection site (100%), which was also transient and mild or moderate
in severity. Evaluation of clinical safety laboratory values grade 2 or higher
and unsolicited adverse events revealed no patterns of concern.

Evaluation of the durability of immune responses is ongoing, and participants
will be followed for one year after the second vaccination, with scheduled blood
collections throughout that period.

Phase 2 Study



We are conducting a Phase 2 placebo-controlled, dose-confirmation study
evaluating the safety, reactogenicity and immunogenicity of two vaccinations of
mRNA-1273 given 28 days apart. Each cohort -- healthy adults ages 18-55 years
(n=300) and older adults ages 55 years and above (n=300) -- is receiving
placebo, a 50 ?g or a 100 ?g dose at both vaccinations. On July 8, 2020, we
announced that the Phase 2 study was fully enrolled. Participants will be
followed for one year after the second vaccination.

Phase 3 Study



We are conducting a Phase 3 randomized, 1:1 placebo-controlled study of
mRNA-1273, named the COVE study, which began enrollment on July 27, 2020. The
study protocol, which has been reviewed by the U.S. Food and Drug Administration
(FDA) and is
                                       50
--------------------------------------------------------------------------------
  Table of Contents
aligned to recent FDA guidance on clinical trial design for COVID-19 vaccine
studies, provides for approximately 30,000 participants in the United States at
the 100 µg dose level. The primary endpoint will be the prevention of
symptomatic COVID-19 disease. Key secondary endpoints include prevention of
severe COVID-19 disease (as defined by the need for hospitalization) and
prevention of infection by SARS-CoV-2. The primary efficacy analysis will be an
event-driven analysis based on the number of participants with symptomatic
COVID-19 disease. The target vaccine efficacy (VE) against COVID-19 for powering
assumptions is 60% (95% confidence interval to exclude a lower bound >30%). Data
will be reviewed by an independent Data Safety Monitoring Board organized by
NIH. The trial is expected to have two interim analyses (at approximately 53 and
106 events), prior to a final event-driven analysis at approximately 151 events.

Regulatory



On May 11, 2020, the FDA granted Fast Track designation for mRNA-1273. Fast
Track is designed to facilitate the development and expedite the review of
therapies and vaccines for serious conditions and fill an unmet medical need.
Programs with Fast Track designation may benefit from early and frequent
communication with the FDA, in addition to a rolling submission of the marketing
application.

Manufacturing

We are continuing to manufacture mRNA-1273 at the Moderna Technology Center, our dedicated manufacturing facility. We have also recently entered into arrangements with third parties to enable larger scale manufacturing and fill-finish capabilities.



In May 2020, we announced a 10-year strategic collaboration agreement with Lonza
Ltd. to enable larger scale manufacture of mRNA-1273 and additional Moderna
products in the future. The companies are establishing manufacturing suites at
Lonza's facilities in the United States and Switzerland for the manufacture of
mRNA-1273 at both sites, and the first batches of mRNA-1273 at Lonza's U.S.
facility were manufactured in July.

In June 2020, we announced a collaboration with Catalent, Inc. for large-scale,
commercial fill-finish manufacturing of mRNA-1273 at Catalent's biologics
facility in Indiana. As part of the agreement, Catalent will provide vial
filling and packaging capacity, as well as additional staffing required for 24x7
manufacturing operations at the site to support production of an initial 100
million doses of the vaccine candidate intended to supply the U.S. market
starting in the third quarter of 2020. Catalent will also provide clinical
supply services from its facilities in Philadelphia, Pennsylvania, including
packaging and labeling, as well as storage and distribution to support our Phase
3 clinical study.

In addition, in July 2020, we announced a collaboration with ROVI for large-scale, commercial fill-finish manufacturing of mRNA-1273 intended in principle to supply markets outside of the United States starting in early 2021 from ROVI's facility in Madrid, Spain.

Key Updates for our Other Development Candidates



•CMV vaccine (mRNA-1647): We completed the third planned interim analysis of
data from the Phase 1 clinical trial of mRNA-1647, which has completed
enrollment and is evaluating the safety and immunogenicity of mRNA-1647 in 181
healthy adult volunteers. The clinical trial population includes those who are
naïve to CMV infection (CMV-seronegative) and those who had previously been
infected by CMV (CMV-seropositive). Participants were randomized to receive
either placebo, or 30, 90, 180 or 300 µg of mRNA-1647 on a dosing schedule of 0,
2 and 6 months. This third planned interim analysis assessed immunogenicity of
the first three dose levels (30, 90, and 180 µg) at twelve months (six months
after the third vaccination). Neutralizing antibody titers (levels of
circulating antibodies that block infection) were assessed in two assays
utilizing epithelial cells and fibroblasts, which measure immune response to the
pentamer and gB vaccine antigens, respectively. gB antigen-specific T cell
responses after the second and third vaccinations were measured in a subset of
CMV-seronegative participants in the 30, 90 and 180 µg dose levels utilizing an
ELISpot assay. Pentamer-specific T cell assays remain in development.
Vaccine-induced neutralizing antibody responses in the CMV-seronegative group
were compared to the baseline neutralizing antibody titers in the
CMV-seropositive group, noting that prior maternal CMV infection is associated
with an approximately 30-fold lower risk of congenital CMV infection compared to
the risk in the setting of maternal primary CMV infection.

In CMV-seronegative participants at twelve months (six months after the third vaccination) in the 30, 90 and 180 µg dose levels:



•A dose-related increase in neutralizing antibody titers was observed in
epithelial cell assays.
•Neutralizing antibody titers against epithelial cell infection were 3.6-fold
and 3.9-fold higher in the 90 and 180 µg dose levels than CMV-seropositive
baseline titers at the 90 and 180 µg dose levels.
•Neutralizing antibody titers against fibroblast infection were 0.7 and 0.9
times the CMV-seropositive baseline titers at the 90 and 180 µg dose levels.
                                       51

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

Table of Contents

In CMV-seropositive participants at twelve months (six months after the third vaccination) in the 30, 90 and 180 µg dose levels:



•A dose-related increase in neutralizing antibody titers was observed in both
epithelial cell and fibroblast assays.
•Neutralizing antibody titers against epithelial cell infection ranged between
14-fold to 31-fold over baseline titers in all dose levels.
•Neutralizing antibody titers against fibroblast infection ranged from 6-fold to
8-fold over baseline titers in all dose levels.

The interim data analysis also included an assessment of safety for the highest
dose level (300 µg). Safety and tolerability data at the 300 µg dose level were
generally similar to that observed at the 180 µg dose level, indicating that the
vaccine was generally well-tolerated. There were no vaccine-related serious
adverse events. The most common solicited local adverse reaction at the 300 µg
dose level was injection site pain. The most common solicited systemic adverse
reactions at the 300 µg dose level were headache, fatigue, myalgia and chills
and for seropositive participants, fever and arthralgia.

•hMPV/PIV3 vaccine (mRNA-1653): The first 10 subjects in the Phase 1b age de-escalation clinical trial of mRNA-1653 have been enrolled and dosed. Further screening and enrollment in this trial is paused due to the COVID-19 pandemic.



•Zika virus vaccine (mRNA-1893): All four cohorts (10 µg, 30 µg, 100 µg, 250 µg)
of the Phase 1 study of mRNA-1893 have been dosed. In April, we announced
positive data from an interim analysis of the 10 µg and 30 µg cohorts. The
clinical trial population includes those who had not been infected by the Zika
virus (flavivirus seronegative) and those who had previously been infected by
the Zika virus (flavivirus seropositive). Participants were randomized to
receive either placebo, 10, 30, 100 or 250 µg of mRNA-1893 on a dosing schedule
of day 1 and day 29. This second planned interim analysis assessed safety and
immunogenicity of the higher dose levels (100 and 250 µg) at day 57, 28 days
after the second vaccination. Neutralizing antibody titers (levels of
circulating antibodies that block infection) were assessed using Plaque
Reduction Neutralizing Test (PRNT50) and microneutralization assays (MN), which
provide equivalent guidance for interpreting the neutralizing immune response.

In the flavivirus-seronegative group:



•Seroconversion rates after the second vaccination reached 100% in the 100 µg
dose level and 98.7% in the 250 µg dose level, based on the PRNT50. MN data were
consistent with PRNT50 data.
•A single vaccination at both the 100 and 250 µg dose levels was sufficient to
seroconvert baseline flavivirus seronegative participants. However, there was a
clear benefit of a two-dose series given 28 days apart.
•Each of the 100 and 250 µg dose levels induced a strong neutralizing Zika
virus-specific antibody response.
•When compared with the 100 µg dose level, the 250 µg dose level did not show a
higher neutralizing antibody response at either Day 29 (after one dose) or Day
57 (after the second dose).

In the flavivirus-seropositive group:



•The percentage of participants achieving a 4-fold boost in pre-existing PRNT50
titers after the second vaccination reached 100% in the 100 µg dose level and
75% in the 250 µg dose level, based on the PRNT50. MN data were generally
consistent with PRNT50 data.

A safety analysis indicated that the 100 and 250 µg dose levels were both
generally well tolerated. There were no vaccine-related serious adverse events.
The most common solicited local adverse reaction was local pain at the injection
site. The most common solicited systemic adverse reactions were headache,
fatigue, myalgia, fever and chills.There was a trend towards more observations
of local erythema and swelling/induration at the injection site with higher dose
levels, in particular after the second vaccine administration, as well as a
trend of more solicited systemic adverse events with the 250 µg dose after the
second administration.

For each of the dose cohorts in the Phase 1 study of mRNA-1893, further analysis of safety and immunogenicity at month 7 and month 13 is pending.



•OX40L (mRNA-2416): Based on available data, earlier this year we decided to
focus the development of mRNA-2416 for the treatment of patients with ovarian
cancer in combination with durvalumab (IMFINZI), a PD-L1 inhibitor. The safety
cohort of the combination arm (mRNA-2416 and durvalumab) of this Phase 1/2
clinical trial continues to enroll, and the Phase 2 dose expansion cohort in
patients with ovarian cancer is actively recruiting participants.

•Antibody against Chikungunya virus (mRNA-1944): We are conducting a Phase 1
dose-escalation study in healthy adults that is randomized and
placebo-controlled. The objective is to evaluate the safety and tolerability of
escalating doses (0.1, 0.3, 0.6, mg/kg dose levels, without dexamethasone
included in the premedication regimen, a dose level cohort at 0.6 mg/kg dose
level, with dexamethasone included in the premedication regimen, with 8 subjects
per cohort) of mRNA-1944 administered
                                       52
--------------------------------------------------------------------------------
  Table of Contents
via intravenous infusion. In addition, there is a dose level cohort in which
subjects were administered two IV infusions of 0.3 mg/kg, one infusion on Day 1
and another subsequent infusion on Day 8, without dexamethasone in the
premedication regimen. No further dose escalation beyond 0.6 mg/kg is planned.
Following a pause due to COVID-19 disruption, enrollment and dosing in this
study resumed, and dosing of all dose level cohorts has been completed.

•Methylmalonic Acidemia (MMA) (mRNA-3704): Enrollment continues to be paused for
this study due to difficulties caused by the pandemic. As a result of the study
pause, the single patient previously enrolled was unable to be dosed in
accordance with study criteria, resulting in de-enrollment of the patient.


Financial Operations Overview

Revenue

To date, we have not recognized any revenue from the sale of potential mRNA medicines. Our revenue has been primarily derived from strategic alliances with Merck, Vertex and AstraZeneca, and from government-sponsored and private organizations including BARDA, DARPA and the Gates Foundation to discover, develop, and commercialize potential mRNA medicines.



Total revenue for the three and six months ended June 30, 2020 was $66.4 million
and $74.7 million, respectively. Total revenue for the three and six months
ended June 30, 2019 was $13.1 million and $29.1 million, respectively. In each
period total revenue was comprised of collaboration revenue and grant revenue.

Collaborative revenue from our strategic alliances as follows (in thousands):
                                  Three Months Ended                           Six Months Ended
                                       June 30,                                    June 30,
                                 2020           2019           2020               2019
Collaboration revenue:
AstraZeneca                   $ 15,884       $    188       $ 17,154       $         1,002
Merck                           10,365          8,659         11,341                19,346
Vertex                           2,193          1,183          4,249                 3,797
    Other                            -              -            155                     -

Total collaboration revenue $ 28,442 $ 10,030 $ 32,899 $ 24,145





Cash received from strategic alliances was $8.1 million and $10.5 million for
the six months ended June 30, 2020 and 2019, respectively. The timing of revenue
recognition is not directly correlated to the timing of cash receipts. Total
deferred revenue related to our strategic alliances as of June 30, 2020 and
December 31, 2019 was $173.0 million and $199.5 million, respectively.

Grant revenue was comprised as follows for the periods presented (in thousands):
                          Three Months Ended                           Six Months Ended
                               June 30,                                    June 30,
                          2020           2019          2020               2019
Grant revenue:
BARDA                 $  37,048       $ 1,876       $ 39,816       $        3,365
Other                       861         1,177          2,025                1,598
Total grant revenue   $  37,909       $ 3,053       $ 41,841       $        4,963




Our ability to recognize revenue from sales of mRNA medicines and become
profitable depends upon our ability to successfully develop and commercialize
mRNA medicines. The rapid acceleration of our work on mRNA-1273 may result in
revenue to us, either based on sales of the product directly or through
collaborators. In addition, we expect to continue to receive funding from our
contract with BARDA, which may result in significant additional amounts of
revenue to Moderna during 2020. To the extent that existing or potential future
strategic alliances generate revenue, our revenue may vary due to many
uncertainties in the development of our mRNA medicines under these strategic
alliances and other factors. We may continue to incur losses for the foreseeable
future, and we expect these losses to increase as we continue our research and
development efforts. We expect our programs to mature and advance
                                       53
--------------------------------------------------------------------------------
  Table of Contents
to later stage clinical development, and we expect expenses to increase as we
seek regulatory approvals for our investigational medicines and begin to
commercialize any approved mRNA medicines.

Research and development expenses



The nature of our business and primary focus of our activities generate a
significant amount of research and development costs.
Research and development expenses represent costs incurred by us for the
following:
•cost to develop our platform;
•discovery efforts leading to development candidates;
•preclinical, nonclinical, and clinical development costs for our programs;
•costs related to pre-launch inventory;
•cost to develop our manufacturing technology and infrastructure; and
•digital infrastructure costs.

The costs above comprise the following categories:
•personnel-related expenses, including salaries, benefits, and stock-based
compensation expense;
•expenses incurred under agreements with third parties, such as consultants,
investigative sites, contract research organizations, or CROs, that conduct our
preclinical studies and clinical trials, and in-licensing arrangements;
•expenses associated with developing manufacturing capabilities and acquiring
materials for preclinical studies, clinical trials and pre-launch inventory,
including both internal manufacturing and third-party contract manufacturing
organizations, or CMOs;
•expenses incurred for the procurement of materials, laboratory supplies, and
non-capital equipment used in the research and development process; and
•facilities, depreciation, and amortization, and other direct and allocated
expenses incurred as a result of research and development activities.
We use our employee and infrastructure resources for the advancement of our
platform, and for discovering and developing programs. Due to the number of
ongoing programs and our ability to use resources across several projects,
indirect or shared operating costs incurred for our research and development
programs are generally not recorded or maintained on a program- or
modality-specific basis. The following table reflects our research and
development expenses, including direct program-specific expenses summarized by
modality and indirect or shared operating costs summarized under other research
and development expenses during the three and six months ended June 30, 2020 and
2019 (in thousands):
                                       54

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


  Table of Contents
                                                         Three Months Ended                                     Six Months Ended
                                                              June 30,                                              June 30,
                                                       2020               2019               2020                  2019
Program expenses by modality:
Prophylactic vaccines                              $  35,657          $  12,398          $  44,397          $        32,660
Cancer vaccines                                       11,652             13,375             16,119                   23,461
Intratumoral immuno-oncology                           1,443              2,775              3,476                    7,193
Localized regenerative therapeutics                        -                  8                  -                       16
Systemic secreted and cell surface therapeutics          578              4,484              1,270                    9,117
Systemic intracellular therapeutics                    5,235              9,829             12,430                   16,572
Total program-specific expenses by modality (1)       54,565             42,869             77,692                   89,019
Other research and development expenses:
Discovery programs                                    10,229             15,635             20,727                   28,550
Platform research                                     18,659             24,256             40,245                   48,753
Technical development and unallocated
manufacturing expenses                                29,284             18,258             58,422                   39,443
Shared discovery and development expenses             24,416             14,355             43,168                   29,238
Stock-based compensation                              14,703             12,932             26,739                   23,715
Total research and development expenses            $ 151,856          $ 

128,305 $ 266,993 $ 258,718

__________


(1)Include a total of 23 and 21 development candidates at June 30, 2020 and
2019, respectively. Program-specific expenses include external costs and
allocated manufacturing costs of pre-launch inventory, mRNA supply and
consumables, and are reflected as of the beginning of the period in which the
program was internally advanced to development or removed if development was
ceased.

A "modality" refers to a group of programs with common product features and the
associated combination of enabling mRNA technologies, delivery technologies, and
manufacturing processes. The program-specific expenses by modality summarized in
the table above include expenses we directly attribute to our programs, which
consist primarily of external costs, such as fees paid to outside consultants,
central laboratories, investigative sites, and CROs in connection with our
preclinical studies and clinical trials, CMOs, and allocated manufacturing costs
of pre-launch inventory, mRNA supply and consumables. Costs to acquire and
manufacture pre-launch inventory, mRNA supply for preclinical studies and
clinical trials are recognized and included in unallocated manufacturing
expenses when incurred, and subsequently allocated to program-specific
manufacturing costs after completion of the program-specific production. The
timing of allocating manufacturing costs to the specific program varies
depending on the program development and production schedule. We generally do
not allocate personnel-related costs, including stock-based compensation, costs
associated with our general platform research, technical development, and other
shared costs on a program-specific basis. These costs were therefore excluded
from the summary of program-specific expenses by modality.

Discovery program expenses are costs associated with research activities for our
programs in the preclinical discovery stage, and primarily consist of external
costs for CROs and lab services, and allocated manufacturing cost of preclinical
mRNA supply and consumables.

Platform research expenses are mainly costs to develop technical advances in
mRNA science, delivery science, and manufacturing process design. These costs
include personnel-related costs, computer equipment, facilities, preclinical
mRNA supply and consumables, and other administrative costs to support our
platform research. Technology development and unallocated manufacturing expenses
are primarily related to non-program-specific manufacturing process development
and manufacturing costs.

Shared discovery and development expenses are research and development costs such as personnel-related costs and other costs, which are not otherwise included in development programs, discovery programs, platform research, technical development and unallocated manufacturing expenses, stock-based compensation, and other expenses.


                                       55
--------------------------------------------------------------------------------
  Table of Contents
The largest component of our total operating expenses has historically been our
investment in research and development activities, including development of our
platform, mRNA technologies, and manufacturing technologies. We expense research
and development costs as incurred and cannot reasonably estimate the nature,
timing, and estimated costs required to complete the development of the
development candidates and investigational medicines we are currently developing
or may develop in the future. There are numerous risks and uncertainties
associated with the research and development of such development candidates and
investigational medicines, including, but not limited to:
•scope, progress, and expense of developing ongoing and future development
candidates and investigational medicines;
•entry in and completion of related preclinical studies;
•enrollment in and completion of subsequent clinical trials;
•safety and efficacy of investigational medicines resulting from these clinical
trials;
•changes in laws or regulations relevant to the investigational medicines in
development;
•receipt of the required regulatory approvals; and
•commercialization, including establishing manufacturing and marketing
capabilities.
A change in expectations or outcomes of any of the known or unknown risks and
uncertainties may materially impact our expected research and development
expenditures. Continued research and development is central to the ongoing
activities of our business. Investigational medicines in later stages of
clinical development, including mRNA-1273 and mRNA-1647, generally have higher
development costs than those in earlier stages of clinical development,
primarily due to the increased size and duration of later-stage clinical trials.
We expect our research and development costs to continue to increase in the
foreseeable future as our investigational medicines progress through the
development phases, as we continue to advance the development of mRNA-1273 and
mRNA-1647 and identify and develop additional programs. There are numerous
factors associated with the successful commercialization of any of our
investigational medicines, including future trial design and various regulatory
requirements, many of which cannot be determined with accuracy at this time due
to the early stage of development of our investigational medicines. Moreover,
future commercial and regulatory factors beyond our control will impact our
clinical development programs and plans.

As we continue to progress mRNA-1273 through the development process in order to
be useful during the current pandemic, we expect to incur significant additional
expenses, including those related to clinical trials, expanding our
manufacturing capabilities, costs of pre-launch inventory, regulatory filings
and the related costs, expansion of our operations into foreign jurisdictions
and commercialization and distribution efforts. At this time, the magnitude of
these potential expenditures and whether or not they will be funded by third
party contributions in whole or in part is not known. In connection with the new
BARDA agreement to accelerate development of mRNA-1273, our revenue and expenses
are expected to increase significantly. BARDA's funding is expected to offset
those increased expenses that are covered under the BARDA agreement, subject to
our obtaining reimbursement from BARDA.

General and administrative expenses



General and administrative expenses consist primarily of personnel-related
costs, including stock-based compensation, for executives, finance, legal, human
resources, business development and other administrative and operational
functions, professional fees, accounting and legal services, information
technology and facility-related costs, and expenses associated with obtaining
and maintaining intellectual property, or IP. These costs relate to the
operation of the business, unrelated to the research and development function,
or any individual program.

We anticipate general and administrative expenses will increase as we continue
to expand the number of programs in development and prepare for the potential
earlier establishment of commercial activities both within and outside the
United States. In addition, if we obtain regulatory approval for any of our
investigational medicines, including the potential accelerated approval for
mRNA-1273, and do not enter into one or more third-party commercialization
collaboration and manufacturing arrangements, we will incur significant expenses
related to building a regulatory, manufacturing, sales and marketing team to
support medicine sales, marketing, and distribution activities.

We have a broad IP portfolio covering our development and commercialization of
mRNA vaccine and therapeutic programs, including those related to mRNA design,
formulation, and manufacturing platform technologies. We regularly file patent
applications to protect innovations arising from our research and development.
We also hold trademarks and trademark applications in the United States and
foreign jurisdictions. Costs to secure and defend our IP are expensed as
incurred and are classified as general and administrative expenses.

                                       56
--------------------------------------------------------------------------------
  Table of Contents
General and administrative expenses, including IP-related expenses, were $36.6
million and $60.7 million for the three and six months ended June 30, 2020,
respectively. General and administrative expenses, including IP-related
expenses, were $28.5 million and $55.7 million for the three and six months
ended June 30, 2019, respectively. IP-related expenses, including our internal
personnel-related costs, were $3.2 million and $5.6 million for the three and
six months ended June 30, 2020, respectively. IP-related expenses, including our
internal personnel-related costs, were $4.4 million and $7.2 million for the
three and six months ended June 30, 2019, respectively. We did not incur
litigation expenses related to our IP during the three months or six months
ended June 30, 2020 and 2019.

Interest income

Interest income consists of interest generated from our investments in cash and cash equivalents, money market funds, and high-quality fixed income securities.

Other expense, net



Other expense, net consists of interest expense, gains (losses) from the sale of
investments in marketable securities, and other income and expense unrelated to
our core operations. Interest expense is primarily derived from our finance
lease related to our Moderna Technology Center manufacturing facility, or MTC
South.

Critical accounting policies and significant judgments and estimates



Our management's discussion and analysis of our financial condition and results
of operations is based on our condensed consolidated financial statements, which
have been prepared in accordance with U.S. generally accepted accounting
principles. The preparation of these condensed consolidated financial statements
requires us to make judgments and estimates that affect the reported amounts of
assets, liabilities, revenues, and expenses and the disclosure of contingent
assets and liabilities in our condensed consolidated financial statements. We
base our estimates on historical experience, known trends and events, and
various other factors that we believe to be reasonable under the circumstances,
the results of which form the basis for making judgments about the carrying
values of assets and liabilities that are not readily apparent from other
sources. Actual results may differ from these estimates under different
assumptions or conditions. On an ongoing basis, we evaluate our judgments and
estimates in light of changes in circumstances, facts, and experience. The
effects of material revisions in estimates, if any, are reflected in the
condensed consolidated financial statements prospectively from the date of
change in estimates.

There have been no material changes in our critical accounting policies and
estimates in the preparation of our condensed consolidated financial statements
during the three months ended June 30, 2020 compared to those disclosed in our
Annual Report on Form 10-K for the year ended December 31, 2019, or 2019 Form
10-K.

Recently issued accounting pronouncements



We have reviewed all recently issued standards and have determined that, other
than as disclosed in Note 2 to our condensed consolidated financial statements,
such standards will not have a material impact on our financial statements or do
not otherwise apply to our operations.
                                       57

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

Table of Contents

Results of operations

The following tables summarize our condensed consolidated statements of operations for each period presented (in thousands):


                                                    Three Months Ended June 30,                                     Change 2020 vs. 2019
                                                      2020                  2019                 $                   %
Revenue:
Collaboration revenue                          $       28,442           $   10,030          $ 18,412               184%
Grant revenue                                          37,909                3,053            34,856               1142%
Total revenue                                          66,351               13,083            53,268               407%
Operating Expenses:
Research and development                              151,856              128,305            23,551                18%
General and administrative                             36,622               28,487             8,135                29%
Total operating expenses                              188,478              156,792            31,686                20%
Loss from operations                                 (122,127)            (143,709)           21,582               (15)%
Interest income                                         7,092               10,322            (3,230)              (31)%
Other expense, net                                     (1,530)              (1,877)              347               (18)%
Loss before income taxes                             (116,565)            (135,264)           18,699               (14)%
Provision for (benefit from) income taxes                 148                 (324)              472              (146)%
Net loss                                       $     (116,713)          $ (134,940)         $ 18,227               (14)%




                                                    Six Months Ended June 30,                                     Change 2020 vs. 2019
                                                     2020                 2019                 $                   %
Revenue:
Collaboration revenue                          $      32,899          $   24,145          $  8,754                36%
Grant revenue                                         41,841               4,963            36,878               743%
Total revenue                                         74,740              29,108            45,632               157%
Operating Expenses:
Research and development                             266,993             258,718             8,275                3%
General and administrative                            60,736              55,740             4,996                9%
Total operating expenses                             327,729             314,458            13,271                4%
Loss from operations                                (252,989)           (285,350)           32,361               (11)%
Interest income                                       14,944              21,294            (6,350)              (30)%
Other expense, net                                    (2,684)             (3,808)            1,124               (30)%
Loss before income taxes                            (240,729)           (267,864)           27,135               (10)%
Provision for (benefit from) income taxes                214                (348)              562              (161)%
Net loss                                       $    (240,943)         $ (267,516)         $ 26,573               (10)%



Revenue

Total revenue increased by $53.3 million, or 407%, for the three months ended
June 30, 2020 compared to the same period in 2019, due to increases in both
collaboration revenue and grant revenue. Collaboration revenue increased by
$18.4 million for the three months ended June 30, 2020 compared to the same
period in 2019, primarily driven by an increase in revenue due to delivery of
drug materials under the collaboration agreements with AstraZeneca. Grant
revenue increased by $34.9 million for the three months ended June 30, 2020
compared to the same period in 2019, mainly due to an increase in revenue from
BARDA related to our mRNA-1273 vaccine candidate development.

Total revenue increased by $45.6 million, or 157%, for the six months ended June
30, 2020 compared to the same period in 2019, due to increases in both
collaboration revenue and grant revenue. Collaboration revenue increased by $8.8
million for the six months ended June 30, 2020 compared to the same period in
2019, mainly attributable to an increase in revenue in the second quarter of
2020, particularly from AstraZeneca, partially offset by cumulative catch-up
adjustments in revenue in the first quarter of 2020 due to changes in estimated
costs for our future performance obligations under the collaboration agreements
with AstraZeneca and Merck.
                                       58
--------------------------------------------------------------------------------
  Table of Contents
Grant revenue increased by $36.9 million for the six months ended June 30, 2020
compared to the same period in 2019, mainly driven by an increase in revenue in
the second quarter of 2020 from BARDA related to our mRNA-1273 vaccine candidate
development.

Operating expenses

Research and development expenses



Research and development expenses increased by $23.6 million, or 18%, for the
three months ended June 30, 2020 compared to the same period in 2019. The
increase was primarily attributable to an increase in personnel-related costs of
$12.4 million and an increase in consulting and outside services of $12.2
million, mainly driven by increased headcount and mRNA-1273 clinical
development. The increases were partially offset by a decrease in raw materials
and manufacturing costs of $4.2 million, mainly due to change in timing of raw
material inventory management and manufacturing lead time.

Research and development expenses increased by $8.3 million, or 3%, for the six
months ended June 30, 2020 compared to the same period in 2019. The increase was
primarily attributable to an increase in personnel related costs of $15.5
million, an increase in consulting and outside services of $11.2 million, and an
increase in stock-based compensation of $3.1 million, largely attributable to
increased headcount and mRNA-1273 clinical development. The increases were
partially offset by a decrease in raw materials and manufacturing costs of $19.4
million and a decrease in lab supplies of $2.8 million, mainly due to change in
timing of raw material inventory management and manufacturing lead time.

General and administrative expenses

General and administrative expenses increased by $8.1 million, or 29%, for the three months ended June 30, 2020 compared to the same period in 2019. The increase was mainly due to an increase in personnel-related costs of $3.6 million and an increase in legal-related costs of $3.6 million.



General and administrative expenses increased by $5.0 million, or 9%, for the
six months ended June 30, 2020 compared to the same period in 2019. The increase
was mainly due to an increase in personnel-related costs of $3.8 million, an
increase in legal-related costs of $2.0 million, and an increase in stock-based
compensation of $1.3 million. The increases were partially offset by a decrease
in consulting and outside services of $2.3 million.

These increases for both the three and six month periods in 2020 were primarily attributable to increased headcount and mRNA-1273 vaccine candidate development-related activities.

Interest income



Interest income decreased by $3.2 million, or 31%, for the three months ended
June 30, 2020 compared to the same period in 2019. Interest income decreased by
$6.4 million, or 30%, for the six months ended June 30, 2020 compared to the
same period in 2019. The decreases in interest income from our investments in
marketable securities for the three and six month periods in 2020 were mainly
attributable to an overall lower interest rate.

Other expense, net



The following table summarizes other expense, net for each period presented (in
thousands):
                                                          Three Months Ended June 30,                                  Change 2020 vs. 2019
                                                            2020                 2019               $                  %
Gain on investments                                   $         570           $     17          $   553              3253%
Interest expense                                             (1,878)            (1,765)            (113)               6%
Other expense, net                                             (222)              (129)             (93)              72%
Total other expense, net                              $      (1,530)          $ (1,877)         $   347              (18)%



                                                           Six Months Ended June 30,                                   Change 2020 vs. 2019
                                                             2020                2019               $                  %
Gain on investments                                    $        891           $     14              877              6264%
Interest expense                                             (3,544)            (3,298)            (246)               7%
Other expense, net                                              (31)              (524)             493              (94)%
Total other expense, net                               $     (2,684)          $ (3,808)         $ 1,124              (30)%


                                       59

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

Table of Contents

Total other expense, net remained relatively flat for the three and six months ended June 30, 2020, compared to the same periods in 2019.

Liquidity and capital resources



We have historically funded our operations primarily from the sale of equity
instruments and from proceeds from certain strategic alliance arrangements and
grant agreements. As of June 30, 2020, we had cash, cash equivalents and
investments of $3.07 billion. Cash, cash equivalents and investments are
invested in accordance with our investment policy, primarily with a view to
liquidity and capital preservation. Investments, consisting primarily of
government and corporate debt securities, are stated at fair value. As of June
30, 2020, we had current and non-current investments of approximately $955.4
million and $354.9 million, respectively.

We began construction of our manufacturing facility in Massachusetts, MTC South,
in the second half of 2016 and completed construction during 2019. In the second
quarter of 2019, we entered into an additional lease for office and laboratory
space nearby, or MTC North. We started construction of MTC North in the fourth
quarter of 2019. Our capital expenditures related to our MTC facilities were
$20.0 million and $3.7 million for the six months ended June 30, 2020 and 2019,
respectively. Cash disbursements related to our MTC facilities were $15.3
million and $10.9 million for the six months ended June 30, 2020 and 2019.

In the second quarter of 2020, we received deposits of $75.0 million for our
future mRNA-1273 vaccine supply based on preliminary agreements with certain of
our potential customers.

Cash flow

The following table summarizes the primary sources and uses of cash for each period presented (in thousands):


                                                                        Six 

Months Ended June 30,


                                                                         2020                 2019
Net cash provided by (used in):
Operating activities                                               $    (130,066)         $ (252,853)
Investing activities                                                    (303,539)           (258,660)
Financing activities                                                   1,959,358               4,470
Net increase (decrease) in cash, cash equivalents and restricted
cash                                                               $   1,525,753          $ (507,043)

Operating activities



We derive cash flows from operations primarily from cash collected from certain
strategic alliances. Our cash flows from operating activities are significantly
influenced by our use of cash for operating expenses and working capital to
support the business. We have historically experienced and will continue to
expect negative cash flows from operating activities due to our investments in
mRNA technologies, digital infrastructure, manufacturing technology and
infrastructure, and advancing our program development efforts and pipeline.

Net cash used in operating activities for the six months ended June 30, 2020 was
$130.1 million and consisted of net loss of $240.9 million and non-cash
adjustments of $61.6 million, plus a net change in assets and liabilities of
$49.3 million. Non-cash items primarily included stock-based compensation of
$44.3 million, and depreciation and amortization of $15.0 million. The net
change in assets and liabilities was due to an increase in deferred revenue of
$51.4 million, an increase in accrued liabilities of $20.2 million, an increase
in operating lease liabilities of $14.0 million, an increase in accounts payable
of $11.5 million and an increase in other liabilities of $4.4 million, partially
offset by an increase in accounts receivable of $28.0 million, an increase in
right-of-use assets related to operating leases of $12.4 million, an increase in
prepaid expenses and an increase in other assets of $11.8 million.

Net cash used in operating activities for the six months ended June 30, 2019 was
$252.9 million and consisted of net loss of $267.5 million and non-cash
adjustments of $52.4 million, minus a net change in assets and liabilities of
$37.8 million. Non-cash items primarily included stock-based compensation of
$40.0 million, depreciation and amortization of $14.8 million and amortization
of investment premium and discount of $2.4 million. The net change in assets and
liabilities was primarily due to a decrease in accrued liabilities of $27.8
million, a decrease in deferred revenue of $23.1 million and an increase in
right-of-use assets relating to operating leases of $3.4 million, partially
offset by a decrease in accounts receivable of $8.1 million, a decrease in
prepaid expense and other assets of $6.0 million, and an increase of
right-of-use assets relating to operating leases of $3.6 million.

                                       60
--------------------------------------------------------------------------------
  Table of Contents
Investing activities

Our primary investing activities consist of purchases, sales, and maturities of
our investments and capital expenditures for manufacturing, laboratory, computer
equipment and software.

Net cash used in investing activities for the six months ended June 30, 2020 was
$303.5 million, which included purchases of marketable securities of $903.6
million and purchases of property and equipment of $24.9 million, partially
offset by proceeds from maturities of marketable securities of $516.9 million
and proceeds from sales of marketable securities of $108.0 million.

Net cash used in investing activities for the six months ended June 30, 2019 was
$258.7 million, which included purchases of marketable securities of $843.3
million and purchases of property and equipment of $18.2 million, partially
offset by proceeds from maturities of marketable securities of $563.6 million
and proceeds from sales of marketable securities of $39.2 million.

Financing activities



We generated cash from financing activities of $1.96 billion for the six months
ended June 30, 2020, primarily from net proceeds from equity offerings of $1.85
billion and net proceeds from the issuance of common stock through our equity
plans of $106.6 million.

We had insignificant financing activities for the six months ended June 30, 2019.

Operation and funding requirements



Since our inception, we have incurred significant losses and negative cash flows
from operations due to our significant research and development expenses. We
have an accumulated deficit of $1.74 billion as of June 30, 2020. We may
continue to incur significant losses in the foreseeable future and expect our
expenses to increase, as we continue research and development of our development
candidates and clinical activities for our investigational medicines. We also
expect our expenses to increase associated with manufacturing costs, pre-launch
inventory expenses, the establishment of late stage clinical and commercial
capabilities, including our arrangements with our international supply and
manufacturing partners. Our ongoing work on mRNA-1273 will require significant
additional investment during 2020, some of which may not be reimbursed or
otherwise paid for by our partners or collaborators. In addition, we expect to
continue to incur additional costs associated with operating as a public company
driven, in part, by the increased compliance requirements of being a publicly
traded company that no longer qualifies as an emerging growth company as of
December 31, 2019.
We are subject to all the risks related to the development and commercialization
of novel medicines, and we may encounter unforeseen expenses, difficulties,
complications, delays, and other unknown factors including the expenses related
to the ongoing coronavirus pandemic, which may adversely affect our business.
Our forecast of the period of time through which our financial resources will be
adequate to support our operations is a forward-looking statement and involves
risks and uncertainties, and actual results could vary as a result of a number
of factors. We have based this estimate on assumptions that may prove to be
wrong, and we could utilize our available capital resources sooner than we
currently expect. We believe that our cash, cash equivalents, and investments as
of June 30, 2020, will be sufficient to enable us to fund our projected
operations through at least the next 12 months from the issuance of our
financial statements.

Until we can generate a sufficient amount of revenue from our programs, we
expect to finance future cash needs through a combination of public or private
equity offerings, structured financings and debt financings, government funding
arrangements, potential future strategic alliances from which we receive upfront
fees, milestone payments, and other forms of consideration, and marketing,
manufacturing, distribution and licensing arrangements. Additional capital may
not be available on reasonable terms, if at all. If we are unable to raise
additional capital in sufficient amounts or on terms acceptable to us, we may
have to significantly delay, scale back, or discontinue the development or
commercialization of one or more of our investigational medicines, or slow down
or cease work on one or more of our programs. If we raise additional funds
through the issuance of additional equity or debt securities, it could result in
dilution to our existing stockholders or increased fixed payment obligations,
and any such securities may have rights senior to those of our common stock. If
we incur indebtedness, we could become subject to covenants that would restrict
our operations and potentially impair our competitiveness, such as limitations
on our ability to incur additional debt, limitations on our ability to acquire,
sell or license intellectual property rights and other operating restrictions
that could adversely impact our ability to conduct our business. If we raise
funds through strategic alliances or marketing, distribution, or licensing
arrangements with third parties, we may have to relinquish valuable rights to
our technologies, future revenue streams, research programs, or investigational
medicines or grant licenses on terms that may not be favorable to us. Any of
these events could significantly harm our business, financial condition, and
prospects.
                                       61

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

Table of Contents

Contractual Obligations



As of June 30, 2020, other than disclosed at Note 7 and Note 8 to our condensed
consolidated financial statements, there have been no material changes to our
contractual obligations and commitments from those described under "Management's
Discussion and Analysis of Financial Condition and Results of Operations"
included in our 2019 Form 10-K.

Off balance sheet arrangements

As of June 30, 2020, we did not have any off-balance sheet arrangements, as defined in Item 303(a)(4)(ii) of Regulation S-K.

© Edgar Online, source Glimpses