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, 2021, which was filed with
the Securities and Exchange Commission (the SEC) on February 25, 2022 (the 2021
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.

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. 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." We have created seven modalities to date:

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

On December 18, 2020, we received an Emergency Use Authorization (EUA) from the
U.S. Food and Drug Administration (FDA) for the emergency use of the Moderna
COVID-19 Vaccine (also referred to as mRNA-1273 and marketed under the brand
name Spikevax) at the 100 µg dose level in individuals 18 years of age or older.
Subsequently, we have also received authorization for our COVID-19 vaccine from
health agencies in more than 70 countries and from the World Health Organization
(WHO). In addition, we have received authorization for a two-dose 100 µg primary
series of our COVID-19 vaccine in adolescents aged 12-17 years in more than 40
countries. We have received authorization for a two-dose 50 µg primary series of
our COVID-19 vaccine in children ages 6 to 11 in more than 35 countries. The
FDA, European Medicines Agency (EMA), Swissmedic and other health agencies
around the world have authorized a booster dose of our COVID-19 vaccine at the
50 µg dose level for adults ages 18 years and older.

Business Highlights and Recent Development

Moderna COVID-19 Vaccine



•Moderna COVID-19 Vaccine (mRNA-1273, Spikevax): In January 2022, we received
full commercial approval for Spikevax to prevent COVID-19 in individuals 18
years of age and older in the United States. Spikevax also has full commercial
approval in individuals 18 years of age and older in Canada and the United
Kingdom, and is approved or authorized in individuals 18 years and older in more
than 70 countries (100 µg dose).

In March 2022, we received approval from the FDA to amend our EUA to allow for a
second booster dose of our COVID-19 vaccine at the 50 µg dose level in adults 50
years of age and older who have received an initial booster of any of the
authorized or approved COVID-19 vaccines and adults 18 years of age and older
with certain kinds of immunocompromise.

•For the first quarter of 2022, we recognized product sales of $5.9 billion from
sales of our COVID-19 vaccine, compared to $1.7 billion in the first quarter of
2021.

•Moderna COVID-19 Vaccine for adolescents and children: In adolescents aged
12-17 years, the primary series (2 dose, 100 µg) of our COVID-19 vaccine is
authorized in more than 40 countries. We made the decision to evaluate the
potential of a two-dose 50 µg primary series to meet regulatory guidance for
immunogenicity in adolescents. We are preparing to submit data for 50 µg
COVID-19 boosters in this age group.
                                       27

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

Table of Contents



In children aged 6-11 years, the primary series (2 dose series, 50 µg) of our
COVID-19 vaccine is authorized in more than 35 countries, including Australia,
Canada and the EU. We are evaluating a 25 µg dose as a primary series and a
booster dose in this age group.

In March 2022, we announced the Phase 2/3 KidCOVE study in children 6 months to
under 6 years successfully met its primary endpoint. This interim analysis
showed a robust neutralizing antibody response in both age groups after a
two-dose 25 µg primary series of mRNA-1273 along with a favorable safety
profile. Based on these data, in April 2022, we submitted an EUA request to the
FDA for authorization of a two-dose 25 ?g primary series of mRNA-1273 for
children 6 months to under 6 years of age. Similar requests are underway with
international regulatory authorities.

Additional Moderna COVID-19 Vaccine Clinical Studies



•Omicron-specific booster candidate (mRNA-1273.529): Our Omicron-specific
booster candidate is being studied to evaluate the immunogenicity, safety and
reactogenicity of mRNA-1273.529 as a single booster dose in adults aged 18 years
and older in two cohorts: individuals who previously received the two-dose
primary series of mRNA-1273 with the second dose being at least six months ago
(cohort 1), or individuals who have received the two-dose primary series and a
50 µg booster dose of mRNA-1273 with the booster dose being at least three
months ago (cohort 2). Participants in both cohorts will receive a single
booster dose of mRNA-1273.529. In the U.S., a Phase 2 study of the
Omicron-specific booster candidate (mRNA-1273.529) as a third or fourth dose is
fully enrolled.

•Beta-specific bivalent booster (mRNA-1273.211): mRNA-1273.211 includes
mutations found in the Beta variant of concern, several of which have been
persistent in more recent variants of concern, including Omicron. A 50 µg
booster dose of mRNA-1273.211 demonstrated superiority compared to a 50 µg
booster dose of mRNA-1273 against Beta, Delta and Omicron variants of concern
one month after administration. Superiority continued six months after
administration for Beta and Omicron variants of concern as well. A 50 µg booster
dose of mRNA-1273.211 was generally well tolerated with a reactogenicity profile
comparable to a booster dose of mRNA-1273 at the 50 µg dose level.

•Omicron-specific bivalent booster candidate (mRNA-1273.214): mRNA-1273.214 is a
bivalent candidate that combines Moderna's Omicron-specific candidate
(mRNA-1273.529) and mRNA-1273. mRNA-1273.214 is being evaluated in a Phase 2/3
study, and the first participant was dosed in March 2022. We expect initial data
on mRNA-1273.214 in June 2022 to inform selection of our candidate for the
Northern Hemisphere fall 2022 booster.

In the United Kingdom, enrollment is ongoing in a Phase 3 to evaluate Omicron-containing candidates as a third or fourth dose in individuals who received any primary series, including COVID-19 vaccinations from other manufacturers.



•Next-generation vaccine candidate against COVID-19 (mRNA-1283): mRNA-1283 is a
next-generation vaccine candidate against COVID-19 and is being developed as a
potential refrigerator-stable mRNA vaccine that will facilitate easier
distribution and administration by healthcare providers. In a Phase 1 study of
mRNA-1283, preliminary results indicate that when administered as primary series
at lower dose levels (10 µg, 30 µg), mRNA-1283 elicits a robust anti-SARS-CoV-2
neutralizing antibody response comparable to the 100 µg mRNA-1273 primary
series. The frequency of local and systemic solicited adverse reactions of the
mRNA-1283 primary series administered at lower dose levels (10 µg, 30 µg) was
overall comparable to mRNA-1273. Enrollment is complete in a Phase 2 study
evaluating booster doses of mRNA-1283, mRNA-1283.211 (SARS-CoV-2/Beta bivalent),
and mRNA-1283.529 (Omicron monovalent).

Other Business Updates



In March 2022, we announced an expansion of our mRNA pipeline with two new
development programs. This announcement reflects our commitment to expanding our
portfolio by building on our experience with our COVID-19 vaccine. The first
development program is a new combination respiratory vaccine candidate
(mRNA-1230) to target three of the most significant viruses causing respiratory
disease in older adults-SARS-CoV-2, influenza and respiratory syncytial virus
(RSV). The second is a program to develop a vaccine candidate (mRNA-1287)
against endemic human coronaviruses (HCoVs). While less-well known than other
coronaviruses, HCoVs are a significant cause of respiratory disease worldwide.
Four HCoVs (HCoV-229E, -NL63, -OC43, and -HKU1) are endemic globally, accounting
for approximately 10% to 30% of upper respiratory tract infections in adults.

In March 2022, we entered into a Memorandum of Understanding with the Government
of the Republic of Kenya to establish Kenya as the location for our mRNA
manufacturing facility, with the assistance of the U.S. Government. We expect to
build a state-of-the-art mRNA facility in Kenya with the goal of producing up to
500 million doses of vaccines each year. We anticipate investing up to $500
million in this new facility which will focus on drug substance manufacturing on
the continent of Africa for the continent of Africa,
                                       28
--------------------------------------------------------------------------------
  Table of Contents
and could also be expanded to include fill/finish and packaging capabilities at
the site. In parallel, we are also working on plans to allow us to fill doses of
our COVID-19 vaccine in Africa as early as 2023, subject to demand.

In March 2022, we executed a strategic partnership agreement with the Australian
Federal Government to establish a state-of-the-art, mRNA vaccine manufacturing
facility in Australia. The facility, when constructed, is expected to provide
people in Australia with access to a domestically manufactured portfolio of mRNA
vaccines against respiratory viruses, including COVID-19, seasonal influenza,
RSV, and other potential respiratory viruses, pending licensure. As part of this
strategic partnership, we expect to support Australia's mRNA research,
development, and industry ecosystem, including engagement with collaborative
research partnerships with Australian institutions and establishing a Regional
Research Center for respiratory medicines and tropical diseases.

In April 2022, we announced plans for a long-term strategic collaboration with
the Government of Canada to establish a state-of-the-art mRNA vaccine
manufacturing facility in Canada. The collaboration is expected to be finalized
following approval of the final agreement by the Government of Canada. The
facility, when constructed, is expected to provide people in Canada with access
to a domestically manufactured portfolio of mRNA vaccines against respiratory
viruses, including COVID-19, seasonal influenza, RSV, and other potential
respiratory viruses, pending licensure. As part of this strategic collaboration,
we expect to support research and development and other commercial
collaborations in Canada.

In March 2022, we announced our global public health strategy through three new
initiatives aimed at advancing mRNA vaccines for the prevention of infectious
diseases. First, we announced a commitment to advance vaccines targeting 15
priority pathogens into clinical studies by 2025. We expect to prioritize HIV,
tuberculosis (TB), malaria, neglected tropical diseases, and priority pathogens
of the WHO and the Coalition for Epidemic Preparedness Innovations (CEPI).
Second, to accelerate research with the aim of advancing additional vaccines, we
are launching a new program, mRNA Access, that will offer researchers use of our
mRNA technology to explore new vaccines against emerging or neglected infectious
disease. Third, we have expanded our pledge to never enforce our patents for
COVID-19 vaccines against manufacturers in 92 low- and middle-income countries
in the Gavi COVAX Advance Market Commitment (AMC), provided that these vaccines
are manufactured solely for use in these countries. We remain willing to license
our technology for COVID-19 vaccines to manufacturers in countries outside of
the AMC 92 on commercially reasonable terms.

Key Updates for our Other Development Candidates



•Seasonal influenza (flu) (mRNA-1010, mRNA-1011, mRNA-1012, mRNA-1020 and
mRNA-1030): As part of our influenza vaccine development strategy, we are
developing five different influenza vaccines. mRNA-1010 is a single
investigational vaccine consisting of four distinct mRNA sequences that encode
the A H1N1, H3N2 and influenza B Yamagata and Victoria lineages in our
proprietary LNP. mRNA-1011 and mRNA-1012 are investigational vaccines that will
include the four WHO-recommended strains and aim to add additional hemagglutinin
(HA) antigens (e.g. H3N2, H1N1). mRNA-1020 and mRNA-1030 are investigational
vaccines that will aim to add neuraminidase (NA) antigens.

In March 2022, an interim analysis of a Phase 2 study of mRNA-1010 identified no
significant safety concerns, and the immunogenicity data is consistent with a
potential for superiority to standard dose vaccine for influenza A strains
(which drives the majority of disease in adults). The interim data is consistent
with potential for non-inferiority to standard dose vaccine in influenza B
strains (primarily a concern in pediatrics). We plan on starting a Phase 3
safety and immunogenicity study of mRNA-1010 in the Southern Hemisphere in in
2022, and are preparing for a Phase 3 efficacy study in fall 2022 if needed.

In April, we announced that the Phase 1 trial of mRNA-1020 and mRNA-1030 dosed
its first participants. The Phase 1/2 randomized, observer-blind, dose-ranging
study will evaluate the safety, reactogenicity and immunogenicity of a single
dose of mRNA-1020 or mRNA-1030 in healthy adults 18 years and older in the U.S.
mRNA-1020 and mRNA-1030 candidates each include eight mRNAs, targeting both
hemagglutinin and neuraminidase at different doses and ratios.

•Respiratory syncytial virus (RSV) vaccine (mRNA-1345): mRNA-1345 is a vaccine
against RSV encoding for a prefusion F glycoprotein, which elicits a superior
neutralizing antibody response compared to the postfusion state. The Phase 1
study of mRNA-1345 to evaluate the tolerability, reactogenicity and
immunogenicity of mRNA-1345 in younger adults, older adults, women of
child-bearing age, older adults of Japanese descent and children is ongoing. All
cohorts are fully enrolled except the RSV seropositive children cohort, which is
ongoing. Dosing in the older adults of Japanese descent (? 60 years) and
children cohorts are ongoing. Phase 1 interim data from the older adult cohort
showed that a single mRNA-1345 vaccination at 50 µg, 100 µg or 200 µg boosted
neutralizing antibody titers against RSV-A by approximately 14-fold and against
RSV-B by approximately 10-fold. The Phase 3 portion of the pivotal global Phase
2/3 study of mRNA-1345 with approximately 34,000 participants is currently
enrolling. The FDA has granted Fast Track designation for mRNA-1345 in adults
older than 60 years of age.

                                       29
--------------------------------------------------------------------------------
  Table of Contents
•HIV vaccines (mRNA-1644 & mRNA-1574): We are currently advancing two HIV
preventative vaccine strategies based on germline targeting and immune-focusing
approaches. Our mRNA-1644 program is designed to test the hypothesis that
sequential administration of priming and boosting HIV immunogens delivered by
mRNA can induce specific classes of B-cell responses and guide their early
maturation toward broadly neutralizing antibody (bnAb) development. The
induction of bnAbs is widely considered to be a goal of HIV vaccination, and
this is the first step in that process. The immunogens being tested in IAVI G002
were developed by scientific teams at IAVI and Scripps Research and will be
delivered via Moderna's mRNA technology. The Phase 1 trial sponsored by IAVI and
supported by the Gates Foundation is ongoing. In addition, we are also advancing
an HIV trimer mRNA vaccine trial of mRNA-1574. The primary hypothesis is that
the soluble and membrane-bound HIV envelope trimer mRNA vaccines will be safe
and well-tolerated by HIV-uninfected individuals and will elicit autologous
neutralizing antibodies. The Phase 1 trial is ongoing and is sponsored and
funded by the Division of AIDS (DAIDS) of the National Institute of Allergy and
Infectious Diseases (NIAID) within the National Institutes of Health (NIH).

•Nipah vaccine (mRNA-1215): We received a safe to proceed from the FDA for a
Phase 1 study of mRNA-1215, our vaccine candidate against the Nipah virus (NiV).
mRNA-1215 was co-developed along with the NIH's Vaccine Research Center and the
Phase 1 clinical testing will be focused on pandemic preparedness.

•Propionic acidemia (PA) (mRNA-3927): The Phase 1/2 clinical trial for
mRNA-3927, our therapy for the treatment of propionic acidemia, or PA, is
ongoing and the first cohort is fully enrolled. We are enrolling other patients
into additional cohorts. All five patients eligible for the Open Label Extension
(OLE) study have elected to participate. The Phase 1/2 study is designed to
evaluate the safety and tolerability of mRNA-3927 in patients with PA. PA, is a
rare, life-threatening, inherited metabolic disorder due to a defect in the
mitochondrial enzyme propionyl-CoA carboxylase (PCC). It primarily affects the
pediatric population. There is no approved therapy for PA, including no approved
enzyme replacement therapy. We have received Rare Pediatric Disease Designation
and Orphan Drug Designation from the FDA and Orphan Drug Designation from the
European Commission for the PA program. The FDA has also granted Fast Track
designation to mRNA-3927. This is the first development candidate to enter the
clinic in our intracellular therapeutics modality.

•Methylmalonic acidemia (MMA) (mRNA-3705): The Phase 1/2 clinical trial for
mRNA-3705, our therapy for the treatment of methylmalonic acidemia, or MMA, is
ongoing and the first cohort has been fully enrolled. The study is now open in
the UK, Canada and the U.S. Moderna is enrolling patients into additional
cohorts. The one patient eligible to participate in the OLE study has elected to
participate. The Phase 1/2 study is designed to evaluate the safety and
tolerability of mRNA-3705 in patients with MMA. MMA is a rare, life-threatening,
inherited metabolic disorder that is primarily caused by a defect in the
mitochondrial enzyme methylmalonyl-coenzyme A mutase, or MUT. It primarily
affects the pediatric population. There is no approved therapy that addresses
the underlying disorder, including no approved enzyme replacement therapy, due
to the complexity of the protein and its mitochondrial localization.

•Glycogen storage disease type 1a (GSD1a) (mRNA-3745): The FDA has granted
mRNA-3745 Orphan Drug Designation and completed its review of the IND
application allowing it to proceed to clinic. Individuals with GSD1a have a
deficiency in glucose-6-phosphatase resulting in pathological blood glucose
imbalance. mRNA-3745 is an IV-administered mRNA encoding human G6Pase enzyme,
designed to restore the deficient or defective intracellular enzyme activity in
patients with GSD1a.

•IL-12 (MEDI1191): AstraZeneca is leading the early clinical development and an
ongoing, open-label multicenter Phase 1 clinical trial of intratumoral
injections of MEDI1191 alone and in combination with the checkpoint inhibitor,
durvalumab. In April 2022, AstraZeneca presented updated Phase 1 data at the
American Association for Cancer Research (AACR) conference. Intratumoral
MEDI1191 combined with durvalumab was safe and the combination showed
preliminary evidence of clinical benefit, with 29% of patients exhibiting
partial responses or stable disease ?12 weeks as best overall response.

•Personalized cancer vaccine (mRNA-4157): Our personalized cancer vaccine, or
PCV, is currently being evaluated in a Phase 1 and Phase 2 study. The
randomized, placebo-controlled Phase 2 study investigating a 1 mg dose of
mRNA-4157 in combination with Merck's pembrolizumab (KEYTRUDA®), compared to
pembrolizumab alone, for the adjuvant treatment of high-risk resected melanoma
is fully enrolled (n=150). The primary endpoint of the Phase 2 study is
recurrence-free survival at 12 months. The Phase 1 in multiple cohorts is
ongoing and the expanded head and neck cohort is recruiting additional patients.
Moderna shares worldwide commercial rights to mRNA-4157 with Merck.


                                       30
--------------------------------------------------------------------------------
  Table of Contents
Our Pipeline

The following chart shows our current pipeline of 46 development programs, grouped by respiratory vaccines, latent & public health vaccines and therapeutics.


                    [[Image Removed: mrna-20220331_g2.jpg]]

Abbreviations: AZ, AstraZeneca; BARDA, Biomedical Advanced Research and
Development Authority; CMV, Cytomegalovirus; DARPA, Defense Advanced Research
Projects Agency; EBV, Epstein-Barr virus; HIV, human immunodeficiency virus;
hMPV, human metapneumovirus; ILCM, Institute for Life Changing Medicines; IL-2,
interleukin 2; IL-12, interleukin 12; IL-23, interleukin 23; IL-36?,
interleukin-36 gamma; NIH, National Institutes of Health; OX40L, wildtype OX40
ligand; RSV, respiratory syncytial virus; VEGF-A, vascular endothelial growth
factor A.
                                       31

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

Table of Contents

We have developed seven modalities, which are summarized as follows:



•Prophylactic vaccines: Our prophylactic vaccines modality currently includes 31
development programs, 21 of which have entered into clinical trials. We have
ongoing Phase 1 trials for our RSV vaccine in pediatrics, flu vaccines
(mRNA-1020 and mRNA-1030), hMPV/PIV3 vaccine (mRNA-1653), EBV vaccine
(mRNA-1189) and HIV vaccines (mRNA-1644 and mRNA-1574). We have ongoing Phase 2
studies for our flu vaccine (mRNA-1010) and Zika vaccine (mRNA-1893). We have
ongoing Phase 3 studies for our RSV vaccine in older adults (mRNA-1345) and CMV
vaccine (mRNA-1647). Our COVID-19 vaccine (mRNA-1273) is described in detail
above. Our ten preclinical programs within our prophylactic vaccines modality
are for a combined COVID-19 and flu vaccine (mRNA-1073), combined COVID-19, flu
and RSV vaccine (mRNA-1230), combined pediatric RSV and hMPV vaccine
(mRNA-1365), pan-HCoV vaccine (mRNA-1278), seasonal flu vaccines (mRNA-1011 and
mRNA-1012), EBV vaccine to prevent long-term sequelae (mRNA-1195), VZV vaccine
(mRNA-1468), HSV vaccine (mRNA-1608) and Nipah vaccine (mRNA-1215). Three other
vaccines as part of public health programs have had positive Phase 1 readouts -
H10N8 vaccine (mRNA-1440), H7N9 flu vaccine (mRNA-1851), and Chikungunya vaccine
(mRNA-1388) - but are not being further developed without government or other
funding.

•Systemic secreted and cell surface therapeutics: We have three systemic
secreted and cell surface therapeutics development candidates in our pipeline.
Our secreted programs include Relaxin (mRNA-0184) for cardiac disorders, PD-L1
(mRNA-6981) for autoimmune hepatitis and IL-2 (mRNA-6231) for autoimmune
disorders. Our IL-2 program (mRNA-6231) is currently in a Phase 1 study, and is
our first autoimmune therapeutic candidate to enter the clinic. The remaining
programs for Relaxin (mRNA-0184) and PD-L1 (mRNA-6981) are currently in
preclinical development. We previously announced positive data from our
Chikungunya Antibody program (mRNA-1944) within this modality; however, we do
not expect to advance our Chikungunya Antibody program without outside funding,
and we are not currently pursuing further development of it at this time.

•Cancer vaccines: We are currently developing three 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, which is fully enrolled. Our second program within
this modality, mRNA-5671, is a KRAS vaccine. We have retained all rights to our
KRAS vaccine from Merck and we are evaluating next steps for the program. Our
third program is our checkpoint vaccine (mRNA-4359), which is in preclinical
studies.

•Intratumoral immuno-oncology: We have two programs in this modality. Our first
program, OX40L/IL-23/IL-36? (Triplet) (mRNA-2752), is currently in a Phase 1
study that is designed as an open-label, multicenter study of intratumoral
injections of Triplet (mRNA-2752) alone or in combination with durvalumab
(anti-PD-L1). Our second program, IL-12 (MEDI1191), is being developed in
collaboration with AstraZeneca. AstraZeneca is currently enrolling an open-label
multicenter Phase 1 clinical trial of intratumoral injections of MEDI1191 alone
and in combination with the checkpoint inhibitor, durvalumab.

•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. We believe these data provide
clinical proof of mechanism for our mRNA technology outside of the vaccine
setting. In 2021, the Phase 2 study met the primary endpoint of safety and
tolerability of AZD8601 for the 3 mg dose. In the study of 11 patients, seven
were treated with AZD8601 VEGF-A mRNA and four received placebo injections.
Numerical trends were observed in endpoints in the heart failure efficacy
domains compared with placebo, including increase in left ventricular ejection
fraction (LVEF) and patient reported outcomes. In addition, all seven patients
treated with AZD8601 had NT-proBNP (a biomarker that measures the level of a
hormone that is elevated in patients with heart failure) levels below heart
failure limit at 6 months follow-up compared to one of four patients treated
with placebo. AstraZeneca has announced that they intend to move AZD8601 into
further studies. Moderna has licensed worldwide commercial rights to AZD8601 to
AstraZeneca.

•Systemic intracellular therapeutics: We have five systemic intracellular
therapeutics development candidates in our pipeline. Our intracellular programs
address propionic acidemia, or PA (mRNA-3927), methylmalonic acidemia (MMA)
(mRNA-3705), phenylketonuria (PKU) (mRNA-3283), glycogen storage disorder type
1a (GSD1a) (mRNA-3745) and Crigler-Najjar Syndrome Type 1 (CN-1) (mRNA-3351). We
have an ongoing Phase 1 clinical trials for PA (mRNA-3927) and MMA (mRNA-3705).
PKU (mRNA-3283), GSD1a (mRNA-3745) and CN-1 (mRNA-3351) are currently in
preclinical development. The FDA has granted Orphan Drug Designation for
mRNA-3745 and has completed its review of the IND application allowing it to
proceed to clinic. We have entered into a collaboration agreement with the
Institute for Life Changing Medicines (ILCM) to license mRNA-3351 to ILCM with
no upfront fees, and without any downstream payments. ILCM will be responsible
for the clinical development of mRNA-3351.

                                       32
--------------------------------------------------------------------------------
  Table of Contents
•Inhaled pulmonary therapeutics: We have one inhaled pulmonary therapeutic
development candidate in our pipeline. Our program addresses cystic fibrosis, or
CF (VXc-522), in collaboration partnership with Vertex Pharmaceuticals. VXc-522
is an mRNA therapeutic designed to treat the underlying cause of CF by enabling
cells in the lungs to produce functional cystic fibrosis transmembrane
conductance regulator (CFTR) protein for the treatment of the 10% of patients
who do not produce any CFTR protein. IND-enabling studies are underway and
Vertex expects to submit an IND for this program in 2022. Moderna has licensed
worldwide commercial rights to VXc-522 to Vertex.

Financial Operations Overview

Revenue



The following table summarizes revenue for the periods presented (in millions):
                                     Three Months Ended March 31,
                                                              2022         2021
Revenue:
Product sales                                               $ 5,925      $ 1,733
Grant revenue                                                   126          194
Collaboration revenue                                            15           10
Total revenue                                               $ 6,066      $ 1,937



For the three months ended March 31, 2022, we recognized $5.9 billion of product
sales from our COVID-19 vaccine, of which $0.9 billion was generated in the
United States and $5.0 billion was generated from the rest of the world. For the
three months ended March 31, 2021, we recognized $1.7 billion of product sales
from our COVID-19 vaccine, of which $1.4 billion was generated in the United
States and $375 million was generated from the rest of the world.

As of March 31, 2022, we had signed supply agreements of approximately
$17.5 billion for the future supply of our COVID-19 vaccine through 2023, based
on the confirmed volume, subject to modifications, and had deferred revenue of
$5.9 billion associated with customer deposits received or billable under these
agreements. In addition, we believe that the SARS-CoV-2 virus will become
endemic in 2022 and as a result, we expect greater seasonality in our product
sales, with sales slightly larger in the second half of 2022 than in the first
half as the Northern Hemisphere enters the fall and winter in connection with
booster sales.

Other than product sales, our revenue has been primarily derived from
government-sponsored and private organizations including BARDA, DARPA and the
Gates Foundation and from strategic alliances with AstraZeneca, Merck and Vertex
to discover, develop, and commercialize potential mRNA medicines.

Grant revenue was comprised as follows for the periods presented (in millions):
                                Three Months Ended March 31,
                                                          2022       2021
Grant revenue:
BARDA (1)                                                $ 122      $ 192
Other                                                        4          2
Total grant revenue                                      $ 126      $ 194


_______
(1) For the three months ended March 31, 2022, $120 million of BARDA grant
revenue was related to our mRNA-1273 program and $2 million was related to our
Zika vaccine program. For the three months ended March 31, 2021, $190 million of
BARDA grant revenue was related to our mRNA-1273 program and $2 million was
related to our Zika vaccine program.

                                       33
--------------------------------------------------------------------------------
  Table of Contents
Collaboration revenue from our strategic alliances was comprised as follows for
the periods presented (in millions):
                                        Three Months Ended March 31,
                                                                   2022      2021
Collaboration revenue:

Merck                                                               10         -
Vertex                                                               4         9
    Other                                                            1         1
Total collaboration revenue                                       $ 15      $ 10



We expect to continue to receive funding from our contract with BARDA. As of
March 31, 2022, the remaining available funding, net of revenue earned under our
agreement with BARDA for the development of our mRNA-1273 vaccine was
$378 million. To the extent that existing or potential future products generate
revenue, our revenue may vary due to many uncertainties in the future product
demand, the development of our mRNA medicines and other factors.

Research and development expenses



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 months ended March 31, 2022 and 2021
(in millions):
                                                                               Three Months Ended
                                                                                   March 31,
                                                                                         2022              2021
Program expenses by modality:
Prophylactic vaccines                                                                 $    190          $    248
Systemic secreted and cell surface therapeutics                                              1                 1
Cancer vaccines                                                                              3                17
Intratumoral immuno-oncology                                                                 6                 6

Systemic intracellular therapeutics                                                          4                 5
Inhaled pulmonary therapeutics (1)                                                           2                 -
Total program-specific expenses by modality (2)                                       $    206          $    277
Other research and development expenses:
Discovery programs                                                                          30                13
Platform research                                                                           34                25
Technical development and unallocated manufacturing expenses                               134                33
Shared discovery and development expenses                                                  130                39
Stock-based compensation                                                                    20                14
Total research and development expenses                                     

$ 554 $ 401

__________


(1)Inhaled pulmonary therapeutics modality was added in the fourth quarter of
2021.
(2)Includes a total of 43 and 30 development candidates at March 31, 2022 and
2021, 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 contract research organizations
(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,
                                       34
--------------------------------------------------------------------------------
  Table of Contents
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.



The largest component of our total operating expenses has historically been our
investment in research and development activities, including preclinical and
clinical development of our product candidates, development of our platform,
mRNA technologies, and manufacturing technologies.

As we continue to pursue our indication expansion of mRNA-1273, and continue to
develop variant-specific COVID-19 vaccine candidates and our next-generation
COVID-19 vaccine candidate, we expect to continue to incur significant
additional expenses. In connection with the BARDA agreement to accelerate
development of mRNA-1273, significant grant revenue and expenses within the
committed funding scope are expected to continue in 2022. BARDA's funding is
expected to offset those expenses that are covered under the BARDA agreement,
subject to our obtaining reimbursement from BARDA. As of March 31, 2022, the
remaining available funding, net of revenue earned was $378 million.

Changes 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, such as our CMV vaccine, RSV vaccine, flu vaccine and our
COVID-19 vaccine, 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 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.

Critical accounting policies and significant judgments and 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 March 31, 2022 compared to those disclosed in our
2021 Form 10-K.
                                       35

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

Table of Contents

Results of operations

The following table summarizes our condensed consolidated statements of income for each period presented (in millions):


                                                 Three Months Ended March 31,                   Change 2022 vs. 2021
                                                    2022                 2021                 $                    %
Revenue:
Product revenue                               $        5,925          $  1,733          $    4,192                242%
Grant revenue                                            126               194                 (68)              (35)%
Collaboration revenue                                     15                10                   5                50%
Total revenue                                          6,066             1,937               4,129                213%
Operating Expenses:
Cost of sales                                          1,017               193                 824                427%
Research and development                                 554               401                 153                38%
Selling, general and administrative                      268                77                 191                248%
Total operating expenses                               1,839               671               1,168                174%
Income from operations                                 4,227             1,266               2,961                234%
Interest income                                           15                 4                  11                275%
Other expense, net                                       (13)              (10)                 (3)               30%
Income before income taxes                             4,229             1,260               2,969                236%
Provision for income taxes                               572                39                 533               1,367%
Net income                                    $        3,657          $  1,221          $    2,436                200%



Revenue

Total revenue increased by $4.1 billion, or 213%, for the three months ended
March 31, 2022, compared to the same period in 2021, mainly due to an increase
in product sales from sales of our COVID-19 vaccine. Product revenue increased
by $4.2 billion, or 242%, for the three months ended March 31, 2022, compared to
the same period in 2021, largely driven by our manufacturing capacity ramp-up.
Grant revenue decreased by $68 million, or 35%, for the three months ended March
31, 2022, compared to the same period in 2021, primarily driven by a decrease in
revenue from BARDA related to our mRNA-1273 vaccine development.

Operating expenses

Cost of sales



Cost of sales for the three months ended March 31, 2022 was $1.0 billion,
including third-party royalties of $207 million. Cost of sales for the three
months ended March 31, 2022 increased by $824 million, or 427%, compared to the
same period in 2021, primarily driven by higher product sales, and consequently
higher third-party royalties and manufacturing costs.

Cost of sales as a percentage of product sales for the three months ended March
31, 2022 was 17%, compared to 11% for the same period in 2021. The increase was
mainly driven by the lack of the pre-launch inventory benefit (that impacted the
first quarter of 2021), coupled with inventory write-downs and a loss on firm
purchase commitments. The increase was partially offset by a favorable customer
mix and the scale up of our manufacturing processes. If inventory sold for the
three months ended March 31, 2021 was valued at cost, our cost of sales for the
period would have been $377 million, or 22%, of our product sales.

We expect our manufacturing costs to increase as we move from a pandemic to a
seasonal market environment for our COVID-19 vaccine. We expect that this shift
will cause our cost of sales for the full year of 2022 to represent a higher
percentage of our product sales.

                                       36
--------------------------------------------------------------------------------
  Table of Contents
Research and development expenses

Research and development expenses increased by $153 million, or 38%, for the
three months ended March 31, 2022, compared to the same period in 2021. The
increase was primarily attributable to an increase in personnel-related costs of
$39 million, an increase in clinical trial expenses of $38 million, an increase
in technology and facility-related costs of $24 million, and an increase in
consulting and outside services of $23 million. These increases for the
three-month period in 2022 were largely driven by increased mRNA-1273 clinical
development and headcount.

We expect that research and development expenses will increase in 2022 as we
continue to progress our indication expansion of mRNA-1273, and continue to
develop our pipeline and advance our product candidates into later-stage
development, in particular our RSV and flu vaccine programs. In addition, we
also expect to incur significant costs related to the development of
variant-specific COVID-19 candidates and our next-generation COVID-19 vaccine
candidate.

Selling, general and administrative expenses



Selling, general and administrative expenses increased by $191 million, or 248%,
for the three months ended March 31, 2022, compared to the same period in 2021.
The increase was mainly due to an endowment to the Moderna Charitable Foundation
(the Foundation) of $50 million, an increase in distributor fees of $47 million,
an increase in personnel-related costs of $31 million, an increase in consulting
and outside services of $29 million, and an increase in marketing expenses of
$15 million. These increases for the three-month period in 2022 were primarily
driven by our COVID-19 vaccine commercialization-related activities, increased
headcount, and the launch of the Foundation.

We expect that selling, general and administrative expenses will increase in
2022, as we continue to build out our global commercial, regulatory, sales and
marketing infrastructure to support the commercialization of our COVID-19
vaccine, and continue to expand the number of programs and our business
operations.

Interest income



Interest income increased by $11 million, or 275%, for the three months ended
March 31, 2022, compared to the same period in 2021. The increase in interest
income from our investments in marketable securities for the three-month period
in 2022 were mainly driven by increased investment balances and an overall
higher interest rate environment.

Other expense, net



The following table summarizes other expense, net for each period presented (in
millions):
                                                         Three Months Ended March 31,                   Change 2022 vs. 2021
                                                            2022                 2021                  $                     %
Loss on investments                                  $            (6)         $      -          $         (6)              100%
Interest expense                                                  (6)               (3)                   (3)              100%
Other expense, net                                                (1)               (7)                    6               (86)%
Total other expense, net                             $           (13)         $    (10)         $         (3)               30%



Total other expense, net increased by $3 million, or 30%, for the three months
ended March 31, 2022, compared to the same period in 2021. The increase in other
expense, net for the three-month period in 2022 was primarily due to realized
losses on available-for-sale debt securities, partially offset by a net gain
related to our balance sheet hedging activities, and foreign currency
transactions and remeasurements. Our interest expense is primarily related to
our finance leases. Please refer to   Note 11   to our condensed consolidated
financial statements.

Income taxes

Provision for income taxes increased by $533 million for the three months ended
March 31, 2022, compared to the same period in 2021, primarily due to an
increase in pre-tax income and a higher effective tax rate in 2022, as 2021
included tax benefit related to the release of the valuation allowance on the
majority of our deferred tax assets. We expect that our effective tax rate will
increase for the full year 2022 compared to 2021, mainly driven by the release
of the valuation allowance on the majority of the deferred tax assets in 2021.
                                       37

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

Table of Contents

Liquidity and capital resources

The following table summarizes our cash, cash equivalents, investments and working capital for each period presented (in millions):


                                  March 31,      December 31,
                                    2022             2021
Financial assets:
Cash and cash equivalents        $   5,048      $       6,848
Investments                          5,067              3,879
Investments, non-current             9,171              6,843
Total                            $  19,286      $      17,570

Working capital:
Current assets                   $  16,350      $      16,071
Current liabilities                  9,238              9,128
Total                            $   7,112      $       6,943



Our 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. Cash, cash equivalents and investments as of March 31,
2022 increased by $1.7 billion, or 10%, compared to December 31, 2021. During
the three months ended March 31, 2022, we generated cash from operations of $2.8
billion, partially offset by repurchases of our common stock of $623 million,
purchases of property and equipment of $132 million, and unrealized losses on
available-for-sale debt securities of $220 million.

Working capital, which is current assets less current liabilities, as of March
31, 2022 increased by $169 million, or 2%, compared to December 31, 2021,
primarily due to a decrease in deferred revenue of $654 million and an increase
in inventory of $501 million, mainly driven by revenue recognized from deferred
revenue in excess of customer deposits received and our increased inventory
levels. The increase was partially offset by a decrease in cash, cash
equivalents and short-term investments of $612 million, primarily due to
purchases of long-term marketable securities.

As of March 31, 2022, we did not have any off-balance sheet arrangements.

Cash flow

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


                                                                     Three 

Months Ended March 31,


                                                                       2022                2021
Net cash provided by (used in):
Operating activities                                               $    2,763          $   2,971
Investing activities                                                   (3,921)              (180)
Financing activities                                                     (642)                26
Net (decrease) increase in cash, cash equivalents and restricted
cash                                                               $   (1,800)         $   2,817



Operating activities

We derive cash flows from operations primarily from cash collected from customer
deposits and accounts receivable related to our COVID-19 vaccine supply
agreements, as well as certain government-sponsored and private organizations
and strategic alliances. Our cash flows from operating activities are
significantly affected by our use of cash for operating expenses and working
capital to support the business.

                                       38
--------------------------------------------------------------------------------
  Table of Contents
In the third quarter of 2020, we entered into supply agreements with the U.S.
Government, other international governments, and Gavi for the supply of our
COVID-19 vaccine and received upfront deposits. As of March 31, 2022, we had
$5.9 billion in deferred revenue related to customer deposits received or
billable. In addition, we expect to continue to receive funding from our
contract with BARDA related to our mRNA-1273 program. As of March 31, 2022, the
remaining available funding from BARDA, net of revenue earned was $378 million.

Net cash provided by operating activities for the three months ended March 31,
2022 was $2.8 billion and consisted of net income of $3.7 billion and non-cash
adjustments of $5 million, partially offset by a net change in assets and
liabilities of $0.9 billion. Non-cash items included deferred income taxes of
$146 million, depreciation and amortization of $79 million, stock-based
compensation of $44 million, and amortization of investment premium and discount
of $18 million. The net change in assets and liabilities was mainly due to a
decrease in deferred revenue of $805 million, an increase in inventory of $501
million, and an increase in prepaid expenses and other assets of $414 million,
partially offset by an increase in income taxes payable of $716 million.

Net cash provided by operating activities decreased by $208 million, or 7%, during the three months ended March 31, 2022, compared to the same period in 2021, primarily attributable to revenue recognized from deferred revenue in excess of customer deposits received, partially offset by increased product sales and higher collection of receivables.

Investing activities

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

Net cash used in investing activities for the three months ended March 31, 2022 was $3.9 billion, which included purchases of marketable securities of $5.6 billion and purchases of property and equipment of $132 million, partially offset by proceeds from sales of marketable securities of $1.4 billion and proceeds from maturities of marketable securities of $441 million.



Net cash used in investing activities increased by $3.7 billion during the three
months ended March 31, 2022, compared to the same period in 2021, primarily
reflecting timing differences related to purchases, sales, and maturities of
marketable debt securities and changes in our portfolio-mix.

Financing activities

Net cash used by financing activities for the three months ended March 31, 2022 was $642 million, primarily from repurchases of common stock of $623 million.



Net cash used in financing activities increased by $668 million during the three
months ended March 31, 2022, compared to the same period in 2021, mainly due to
repurchases of common stock.

Operation and funding requirements



Our principal sources of funding as of March 31, 2022 consisted of cash and cash
equivalents, investments, and cash we expect to generate from operations. We
generated net income of $12.2 billion for the year ended 2021, following the
authorization of our first commercial product in December 2020. From our
inception to the end of 2020, we incurred significant losses from operations due
to our significant research and development expenses. We have retained earnings
of $13.6 billion as of March 31, 2022.

We have significant future capital requirements including expected operating
expenses to conduct research and development activities, operate our
organization, meet capital expenditure needs, and fund our share repurchase
program (refer to   Note 13   to our condensed consolidated financial
statements). We expect our expenses to increase in connection with our ongoing
activities, particularly 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, including our arrangements with our international supply and
manufacturing partners. Our ongoing work on mRNA-1273, including development of
any new generations of boosters and vaccines against variants of SARS-CoV-2, and
buildout of global commercial, regulatory, sales and marketing infrastructure to
support the commercialization of our COVID-19 will require significant cash
outflows during 2022, most of which may not be reimbursed or otherwise paid for
by our partners or collaborators. In addition, we have substantial facility,
lease and purchase obligations (refer to   Note 11   and   Note 12   to our
condensed consolidated financial statements). We have entered into certain
collaboration agreements with third parties that include the funding of certain
research and development activities and potential future milestone and royalty
payments by us.

We believe that our cash, cash equivalents, and investments as of March 31, 2022, together with cash expected to be generated from operations, will be sufficient to enable us to fund our projected operations, capital expenditures and stock repurchases through at least


                                       39
--------------------------------------------------------------------------------
  Table of Contents
the next 12 months from the issuance of these financial statements included in
this Form 10-Q. 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
expenses related to the ongoing COVID-19 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.

If we fail to sustain profitability on a continuing basis, we may be required 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. If we are required to
finance future cash needs, 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.

Contractual Obligations

As of March 31, 2022, other than disclosed within Note 11 and Note 12

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 2021 Form 10-K.

© Edgar Online, source Glimpses