Forward-looking Information



This Quarterly Report on Form 10-Q contains forward-looking statements which are
made pursuant to the safe harbor provisions of Section 27A of the Securities Act
of 1933, as amended, and Section 21E of the Securities Exchange Act of 1934, as
amended (the "Exchange Act"). These statements may be identified by such
forward-looking terminology as "may," "should," "expects," "intends," "plans,"
"anticipates," "believes," "estimates," "predicts," "potential," "continue" or
the negative of these terms or other comparable terminology. Our forward-looking
statements are based on a series of expectations, assumptions, estimates and
projections about our company, are not guarantees of future results or
performance and involve substantial risks and uncertainty. We may not actually
achieve the plans, intentions or expectations disclosed in these forward-looking
statements. Actual results or events could differ materially from the plans,
intentions and expectations disclosed in these forward-looking statements. Our
business and our forward-looking statements involve substantial known and
unknown risks and uncertainties, including the risks and uncertainties inherent
in our statements regarding:


our ability to execute our clinical study strategy for NTLA-2001, our program
for the treatment of transthyretin ("ATTR") amyloidosis, including the ability
to successfully complete our Phase 1 study and determine a recommended dose in
our ongoing Phase 1 study that can be advanced into later-stage studies, or the
success of such program;


our ability to execute our clinical study strategy for NTLA-2002, our program
for the treatment of hereditary angioedema ("HAE"), including the ability to
successfully complete our Phase 1/2 study and determine a recommended dose that
can be advanced into later-stage studies, or the success of such program;

the anticipated timing of our Investigational New Drug ("IND") or IND-equivalent filing for NTLA-3001, our program for the treatment of alpha-1 antitrypsin deficiency ("AATD")-associated lung disease, or the success of such program;

our ability to successfully execute our development plans for our preclinical programs, including NTLA-2003 and NTLA-6001;

our ability to use a modular platform capability or other strategies to efficiently discover and develop product candidates, including by applying learnings from one program to other programs;

our ability to research, develop or maintain a pipeline of product candidates, including in vivo and allogeneic ex vivo product candidates;

our ability to manufacture or obtain materials for our preclinical and clinical studies, and our product candidates;


our ability to advance any product candidates into, and successfully complete,
clinical studies, including clinical studies necessary for regulatory approval
and commercialization, and to demonstrate to the regulators that the product
candidates are safe, effective, pure and potent and that their benefits outweigh
known and potential risks for the intended patient population;

our ability to advance our genome editing and therapeutic delivery capabilities;

the scope of protection we are able to develop, establish and maintain for intellectual property rights, including patents and license rights, covering our product candidates and technology;

our ability to operate, including commercializing products, without infringing or breaching the proprietary or contractual rights of others;


the issuance or enforcement of, and compliance with, regulatory requirements and
guidance regarding preclinical and clinical studies relevant to genome editing
and our product candidates;

the market acceptance, pricing and reimbursement of our product candidates, if approved;

estimates of our expenses, future revenues, capital requirements and our needs for additional financing;

the potential benefits of strategic agreements, such as collaborations, co-development and co-commercialization, acquisitions, dispositions, mergers, joint ventures, and investment agreements, and our ability to establish and maintain strategic arrangements under favorable terms;


                                       23
--------------------------------------------------------------------------------

our ability to acquire and maintain relevant intellectual property licenses and rights, and the scope and terms of such rights;


developments relating to our licensors, licensees, third parties and ventures
from which we derive or license rights, as well as collaborators, competitors
and our industry;

the effect of the ongoing COVID-19 pandemic, including mitigation efforts and economic effects, on any of the foregoing or other aspects of our business operations; and

other risks and uncertainties, including those listed under the caption "Risk Factors."



All of our express or implied forward-looking statements are as of the date of
this Quarterly Report on Form 10-Q only. In each case, actual results may differ
materially from such forward-looking information. We can give no assurance that
such expectations or forward-looking statements will prove to be correct. An
occurrence of or any material adverse change in one or more of the risk factors
or risks and uncertainties referred to in this Quarterly Report on Form 10-Q or
included in our other public disclosures or our other periodic reports or other
documents or filings filed with or furnished to the Securities and Exchange
Commission (the "SEC") could materially and adversely affect our business,
prospects, financial condition and results of operations. Except as required by
law, we do not undertake or plan to update or revise any such forward-looking
statements to reflect actual results, changes in plans, assumptions, estimates
or projections or other circumstances affecting such forward-looking statements
occurring after the date of this Quarterly Report on Form 10-Q, even if such
results, changes or circumstances make it clear that any forward-looking
information will not be realized. Any public statements or disclosures by us
following this Quarterly Report on Form 10-Q that modify or impact any of the
forward-looking statements contained in this Quarterly Report on Form 10-Q will
be deemed to modify or supersede such statements in this Quarterly Report on
Form 10-Q.

Management Overview

Intellia Therapeutics, Inc. ("we," "us," "our," "Intellia," or the "Company") is
a leading clinical-stage genome editing company, focused on developing novel,
potentially curative therapeutics leveraging CRISPR/Cas9-based technologies.
CRISPR/Cas9, an acronym for Clustered, Regularly Interspaced Short Palindromic
Repeats ("CRISPR")/CRISPR associated 9 ("Cas9"), is a technology for genome
editing, the process of altering selected sequences of genomic deoxyribonucleic
acid ("DNA"). To fully realize the transformative potential of CRISPR/Cas9-based
technologies, we are building a full-spectrum genome editing company, by
leveraging our modular platform, to advance in vivo and ex vivo therapies for
diseases with high unmet need by pursuing two primary approaches. Our in vivo
programs use intravenously administered CRISPR as the therapy, in which
proprietary delivery technology enables highly precise editing of
disease-causing genes directly within specific target tissues. Our ex vivo
programs use CRISPR to create the therapy by using engineered human cells. Our
deep scientific, technical and clinical development experience, along with our
robust intellectual property ("IP") portfolio, have enabled us to take a
leadership role in harnessing the full potential of genome editing to create new
classes of genetic medicine.

Our management's discussion and analysis of our financial condition and results
of operations are based upon our unaudited condensed consolidated financial
statements included in this Quarterly Report on Form 10-Q, which have been
prepared by us in accordance with accounting principles generally accepted in
the United States of America ("U.S. GAAP") for interim periods and with
Regulation S-X, promulgated under the Securities Exchange Act of 1934, as
amended. This discussion and analysis should be read in conjunction with the
unaudited condensed consolidated financial statements and the notes thereto
included elsewhere in this Quarterly Report on Form 10-Q as well as in
conjunction with the audited financial statements and notes thereto included in
our Annual Report on Form 10-K ("Annual Report") for the year ended December 31,
2021.

Treating-and potentially curing-a broad range of severe diseases will require
multiple gene editing approaches. With proprietary CRISPR/Cas9-based technology
at the core of our platform, we continue to add new capabilities to expand our
current solutions for addressing a multitude of life-threatening diseases. These
additions include our proprietary base editor, as well as novel CRISPR enzymes,
which provide us with the capabilities to achieve multiple editing strategies.

We continue to advance our platform's modular solutions and research efforts on genome editing technologies as well as delivery and cell engineering capabilities to generate additional development candidates.



Our mission is to transform the lives of people with severe diseases by
developing curative genome editing treatments. We believe we can deliver on our
mission and provide long-term benefits for all of our stakeholders by focusing
on four key elements:

Develop curative CRISPR/Cas9-based medicines;


                                       24
--------------------------------------------------------------------------------



•
Advance our science;

•

Be the best place to make therapies; and

Focus on long-term sustainability.



Our strategy is to build a full-spectrum genome editing company, by leveraging
our modular platform, to advance in vivo and ex vivo therapies for diseases with
high unmet need. For in vivo applications to address genetic diseases, we deploy
CRISPR/Cas9 as the therapy that targets cells within the body. In parallel, we
are developing ex vivo applications to address immuno-oncology and autoimmune
diseases, where CRISPR/Cas9 is the tool that creates the engineered cell
therapy. All of our revenue to date has been collaboration revenue. Since our
inception and through June 30, 2022, we have raised an aggregate of
approximately $1,861.1 million to fund our operations through our initial public
offering ("IPO") and concurrent private placements, follow-on public offerings,
at-the-market offerings, and the sale of convertible preferred stock, as well as
through our collaboration agreements.

Our lead in vivo candidate, NTLA-2001 for the treatment of transthyretin
("ATTR") amyloidosis, is the first CRISPR/Cas9-based therapy candidate to be
administered systemically, via intravenous infusion, for precision editing of a
gene in a target tissue in humans. In parallel, we are developing ex vivo
applications to address immuno-oncology and autoimmune diseases, where
CRISPR/Cas9 is the tool that creates the engineered cell therapy. For NTLA-5001,
a wholly-owned, autologous T cell receptor ("TCR")-T cell candidate for the
treatment of acute myeloid leukemia ("AML"), we are pivoting towards an
allogeneic version of this program currently in preclinical development. In
addition, Novartis Institutes for BioMedical Research, Inc. ("Novartis") is
advancing a clinical program using our ex vivo technology to engineer
hematopoietic stem cells ("HSCs") for the treatment of sickle cell disease.

Our Pipeline

In Vivo Programs



Our selection criteria include identifying diseases that originate in the liver;
have well-defined mutations that can be addressed by a knockout or insertion
approach; have readily measurable therapeutic endpoints with observable clinical
responses; and for which effective treatments are absent, limited or unduly
burdensome. Our initial in vivo indications target genetic liver diseases,
including our ATTR amyloidosis, hereditary angioedema ("HAE") and alpha-1
antitrypsin deficiency ("AATD") development programs. Our current efforts on in
vivo delivery focus on the use of lipid nanoparticles ("LNPs") for delivery of
the CRISPR/Cas9 complex to the liver.

Transthyretin ("ATTR") Amyloidosis Program



NTLA-2001 is the first investigational CRISPR-based therapy to be systemically
delivered to edit genes inside the human body and has the potential to be the
first single-dose treatment for ATTR amyloidosis. Delivered with our in vivo LNP
technology, NTLA-2001 offers the possibility of halting and reversing the
disease by driving a deep, potentially lifelong reduction in transthyretin
("TTR") protein after a single dose. NTLA-2001 is being evaluated in a Phase 1,
two-part, open-label study in adults with hereditary transthyretin amyloidosis
with polyneuropathy ("ATTRv-PN") or transthyretin amyloidosis with
cardiomyopathy ("ATTR-CM"). To date, over 30 patients have been treated across
the polyneuropathy and cardiomyopathy arms. The growing body of data,
particularly at the two highest doses tested, demonstrated treatment with
NTLA-2001 resulted in rapid, deep and highly consistent reductions of serum TTR.

In the ATTR-CM arm, in August 2022, we announced the completion of the
dose-escalation portion of the study. We are finalizing selection of a fixed
dose, at or near the fixed-dose equivalent of the 0.7 mg/kg dose, for evaluation
in the dose-expansion portion of the study, subject to regulatory approval. The
selection is based on clinical data from patients with ATTR-CM dosed at the 0.7
mg/kg and 1.0 mg/kg doses which yielded similar TTR reductions. Additionally,
both doses were generally well-tolerated. We remain on track to present interim
data from the cardiomyopathy arm later this year.

In the ATTRv-PN arm, in August 2022, subject to regulatory approval, we
announced plans to add a second cohort to the dose-expansion portion of the
polyneuropathy arm, which will evaluate the same fixed dose selected for the
dose-expansion portion of the cardiomyopathy arm cohort. The decision to study a
second dose is based on the following: (1) the emerging data from the
dose-escalation portion of the cardiomyopathy arm showed similar serum TTR
reduction at both the 0.7 mg/kg and 1.0 mg/kg doses, (2) the comparability of
performance at the 0.7 mg/kg and 1.0 mg/kg dose in the dose-escalation portion
of the polyneuropathy arm, which led to an 86% and 93% mean and 97% and 98%
maximum TTR reduction at day 28, respectively,

                                       25
--------------------------------------------------------------------------------


and (3) a significant elevation in liver enzymes, which normalized without
medical intervention, observed at day 28 in a patient treated in the
dose-expansion portion of the polyneuropathy arm at the 80 mg dose (the fixed
dose corresponding to 1.0 mg/kg). While the adverse event is considered possibly
related to study drug, this patient was asymptomatic, had no increase in
bilirubin and the event was deemed nonserious by the investigator. We plan to
submit a protocol amendment to evaluate a fixed dose corresponding to 0.7 mg/kg
in the dose-expansion portion, with enrollment across both arms expected to be
completed by the end of 2022, subject to regulatory feedback.

In June 2022, we presented at the European Association for the Study of the
Liver ("EASL") International Liver Congress 2022 updated interim data from our
ongoing Phase 1 study of NTLA-2001 in patients with ATTRv-PN. Extended follow-up
data from 15 ATTRv-PN patients, treated across all four single-ascending dose
cohorts, showed deep, dose-dependent reductions in serum TTR observed with prior
readouts were sustained through the last measured timepoint of follow-up,
reaching 12 months in the 0.1 mg/kg and 0.3 mg/kg cohorts and six months in the
0.7 mg/kg and 1.0 mg/kg cohorts. Both 0.7 mg/kg and 1.0 mg/kg doses led to
greater than 85% mean TTR reduction at day 28. The durability and persistence of
effect continue to support NTLA-2001 as a potential one-time treatment to
permanently inactivate the TTR gene and reduce the disease-causing protein.

NTLA-2001 is the subject of a co-development and co-promotion ("Co/Co")
agreement directed to our first collaboration target with Regeneron
Pharmaceuticals, Inc. ("Regeneron"), ATTR (the "ATTR Co/Co"), for which we are
the clinical and commercial lead party and Regeneron is the participating party.
Regeneron shares in approximately 25% of worldwide development costs and
commercial profits for the ATTR program. For more information regarding our
collaboration with Regeneron, see the section below entitled "Collaborations and
Other Arrangements - Regeneron".

Hereditary Angioedema ("HAE") Program



NTLA-2002 is our wholly-owned candidate for the treatment of HAE. We are
progressing the single-ascending dose portion of our first-in-human study. We
anticipate presenting interim data in the second half of 2022, including safety,
kallikrein reduction and HAE attack rate data. These initial results are
expected to characterize the emerging safety and activity profile of NTLA-2002,
and potentially demonstrate the modularity of our proprietary, CRISPR-based, LNP
platform.

Alpha-1 Antitrypsin Deficiency ("AATD") Program

NTLA-3001 for associated lung disease:



NTLA-3001 is our wholly-owned, first-in-class CRISPR-mediated in vivo targeted
gene insertion development candidate for the treatment of AATD-associated lung
disease. It is designed to precisely insert a functional copy of the SERPINA1
gene, which encodes the alpha-1 antitrypsin ("A1AT") protein, with the potential
to restore permanent expression of functional A1AT protein to therapeutic levels
after a single dose. This approach seeks to improve patient outcomes, including
eliminating the need for weekly IV infusions of A1AT augmentation therapy or
lung transplant in severe cases. We are conducting investigational new drug
("IND")-enabling activities for NTLA-3001, with plans to file an IND or
IND-equivalent in 2023.

NTLA-2003 for associated liver disease:



NTLA-2003 is our wholly-owned in vivo knockout development candidate for the
treatment of AATD-associated liver disease. It is designed to inactivate the
SERPINA1 gene responsible for the production of abnormal A1AT protein in the
liver. This approach aims to halt the progression of liver disease and eliminate
the need for liver transplant in severe cases. We are conducting IND-enabling
activities for NTLA-2003.

In Vivo Research Programs

We continue to work on various liver-focused programs, such as hemophilia A and
hemophilia B, which we are co-developing with Regeneron, primary hyperoxaluria
type 1, as well as other liver targets, which are worked on both independently
and in partnership with Regeneron, which leverage our capabilities to knockout,
insert and make consecutive edits to the genome.

In the third quarter of 2021, we and Regeneron, the lead party for this program,
nominated a Factor 9 ("F9") gene insertion development candidate for our
Hemophilia B ("Hem B") program, leveraging our jointly developed targeted
transgene insertion capabilities to insert F9. F9 is a gene that encodes for
Factor IX ("FIX"), a blood-clotting protein that is missing or defective in Hem
B patients. In preclinical studies, we and Regeneron demonstrated the first
CRISPR/Cas9-mediated targeted transgene insertion in the liver of non-human
primates ("NHPs"), which resulted in circulating FIX levels at or above those
found in normal human plasma. At the 2019 American Society of Gene and Cell
Therapy Annual Meeting, we presented data demonstrating the

                                       26
--------------------------------------------------------------------------------


first CRISPR/Cas9-mediated, targeted transgene insertion in the liver of NHPs,
using F9 as a model gene. Following a single dose to NHPs of the hybrid
LNP-adeno-associated virus ("AAV") delivery system containing an F9 DNA
template, we demonstrated that the circulating human FIX protein levels achieved
in NHPs were at or above normal levels. Additionally, the NHP data expands on
the durability of clinically relevant human FIX protein levels achieved in mice
for over 12 months.

In September 2020, we presented data that showed the persistence of in vivo
CRISPR/Cas9 edits in regenerated liver tissue, both knockout and insertion, and
corresponding durability of effect following a partial hepatectomy ("PHx") and
liver regrowth in a murine model. Unlike traditional gene therapy, for which a
significant loss (over 80%) in transgene expression was observed in the
insertion PHx model, our targeted gene insertion approach yielded durable edits,
with no significant loss in expression.

We are further investigating delivery strategies that target tissues outside of
the liver. For example, at the Keystone eSymposium: Precision Engineering of the
Genome, Epigenome and Transcriptome in March 2021, we presented preclinical data
establishing proof-of-concept for non-viral genome editing of bone marrow and
HSCs in mice. This represented our first demonstration of systemic in vivo
genome editing in bone marrow using our proprietary non-viral delivery platform.
We believe these results extend our modular in vivo capabilities to treat
inherited blood disorders such as sickle cell disease. In addition, we announced
a collaboration with SparingVision SAS ("SparingVision") to develop novel
genomic medicines utilizing CRISPR/Cas9 technology for the treatment of ocular
diseases.

With the continued progression of our in vivo research programs, we plan to advance at least one new in vivo development candidate by the end of 2022.

Ex Vivo Programs



We are independently researching and developing proprietary engineered cell
therapies to treat various oncological and other disease indications, for
example TCR-engineered T cells and chimeric antigen receptor T ("CAR-T") cells
for immuno-oncology applications and engineered regulatory T cells for
autoimmune disorders. Our diverse product strategy includes multiple elements.
In particular:


We are developing TCR-engineered T cells as immuno-oncological therapies. For
example, in our existing collaboration with Ospedale San Raffaele, Milan, a
leading European research-university hospital, we have identified optimized TCRs
that recognize a tumor target, Wilms' Tumor 1 ("WT1"), that could be used to
treat a variety of blood cancers and solid tumors; and


We are developing allogeneic cellular therapies, which are those derived from
unrelated donors and modified outside of the human body to allow them to be
administered to an unrelated patient. These allogeneic cellular therapies could
be used to treat both oncological and immunological diseases. Our proprietary
technologies, including our LNP-based cell engineering platform and novel
allogeneic solution, offer significant advantages over both autologous cell
therapies and allogeneic approaches being investigated by others. Preclinical
data presented on our differentiated allogeneic engineering platform showed
allogeneic T cells were shielded from immune rejection, both host T and natural
killer ("NK") cell attack.

In addition, we strategically partner with others who possess complementary
capabilities or technologies to bring forth innovative engineered cell therapies
outside of our core areas of focus. This includes collaborations with AvenCell
and Kyverna, who will be leveraging our ex vivo genome editing platform to
develop novel cell therapies for a variety of therapeutic indications, as well
as ONK Therapeutics, Ltd. ("ONK") to advance CRISPR-edited NK cell therapies.
Further, our partner Novartis is developing therapies directed to selected
targets using CAR-T cells for oncology indications, as well as HSC and ocular
stem cell ("OSC")-based therapies.

Acute Myeloid Leukemia ("AML") Program

NTLA-5001 is our investigational autologous TCR-T cell therapy engineered to target the WT1 antigen for the treatment of all genetic subtypes of AML. In March 2022, we dosed the first patient in our Phase 1/2a study evaluating NTLA-5001 for the treatment of AML.



In August 2022 we announced our decision to concentrate our ex vivo development
efforts exclusively on allogeneic cell therapies manufactured from healthy
donors. Our proprietary technologies, including our LNP-based cell engineering
platform and novel allogeneic solution, offer significant advantages over both
autologous and current investigational allogeneic approaches. As

                                       27
--------------------------------------------------------------------------------

described below under CD30+ Lymphomas, preclinical data presented on our differentiated allogeneic engineering platform showed allogeneic T cells were shielded from immune rejection, both host T and NK cell attack.



In addition, we announced plans to discontinue our first-in-human study of
NTLA-5001 and are pivoting to an allogeneic version of this program currently in
preclinical development. This decision is not due to any safety or efficacy data
emerging from the trial. It is instead based on the potential of our allogeneic
platform to consistently deliver a high-quality, readily available and
persistent cell product for treatment of aggressive cancers. Preclinical data
supporting the development of a WT1-directed allogeneic TCR-T cell candidate
will be presented at a future scientific conference in 2022.

CD30+ Lymphomas

NTLA-6001 is our wholly-owned, allogeneic CAR-T development candidate targeting CD30 for the treatment of CD30-expressing hematologic cancers, including relapsed or refractory classical Hodgkin lymphoma ("cHL"). NTLA-6001 is the first candidate developed using our proprietary allogeneic cell engineering platform. We are conducting IND-enabling activities for NTLA-6001.



At the Keystone Symposium on May 1, 2022 we presented preclinical data leading
to the development of NTLA-6001. The data demonstrated that our proprietary
allogeneic solution created T cells that not only avoid immune recognition by
host CD4 and CD8 T cells, but also were protected from NK cell-mediated killing
in in vitro and in vivo mouse models. Furthermore, allogeneic T cells engineered
specifically with LNPs retained their viability, cell expansion, memory
phenotype, cytotoxic and cytokine secretion characteristics.

Ex Vivo Research Programs



We are developing engineered cell therapies to treat a range of hematological
and solid tumors. We are pursuing modalities, such as TCR, with broad potential
in multiple indications. We are advancing efforts for allogeneic therapies to
move from liquid to solid tumors. Our researchers are developing and improving
cell-engineering manufacturing and delivery processes that, we believe, may
allow us to deliver T cell therapies with high levels of editing, robust levels
of cell expansion, desirable memory phenotypes, improved function and no
translocations above background levels.

Our proprietary T cell engineering process using LNPs to engineer cell therapies
enables multiple, sequential gene edits. We have shared preclinical data
demonstrating that our LNP-based engineering technology is a significant
improvement over electroporation, the standard engineering process used to
introduce proteins and nucleic acids into cells. The resulting T cells
engineered with LNPs had improved cell properties and performance both in vitro
and in vivo as compared to electroporation. The data support the ability of our
platform to be used for a variety of targeting modalities, including CAR and
TCRs, and to support both autologous and allogeneic T cell candidates. The
LNP-based approach has been used in our NTLA-5001 program and multiple ex vivo
candidates in development by us and our collaborators.

In March 2021, we presented our first preclinical data set on our novel, proprietary cytosine deaminase base editor technology. We demonstrated the technology's potential for enhanced cell engineering, with multiple simultaneous gene knockouts achieving >90% T cell editing efficiency and no detectable increase in translocation above background levels.

Novartis-Led Sickle Cell Disease and Other Research Programs



In December 2019, the research term under our collaboration agreement with
Novartis entered into in 2014 (the "2014 Novartis Agreement") ended, although
the 2014 Novartis Agreement remains in effect. Under the 2014 Novartis
Agreement, Novartis has selected particular CAR-T cell, HSC and OSC targets for
continued development. Novartis has initiated clinical studies for OTQ923 and
HIX763, two therapeutic candidates, based on CRISPR/Cas9 editing of HSCs that
resulted from our research collaboration with Novartis. Novartis is currently
recruiting patients for its Phase 1/2 study of these investigational candidates
for treatment of sickle cell diseases. Novartis is developing several other
product candidates arising from the 2014 Novartis Agreement. For more
information regarding our collaboration with Novartis, see the section below
entitled "Collaborations and Other Arrangements - Novartis".

                                       28
--------------------------------------------------------------------------------

Collaborations and Other Arrangements



To accelerate the development and commercialization of CRISPR/Cas9-based
products in multiple therapeutic areas, we have formed, and intend to seek other
opportunities to form, strategic alliances with collaborators who can augment
our leadership in CRISPR/Cas9 therapeutic development.

Regeneron



In April 2016, we entered into a license and collaboration agreement with
Regeneron (the "2016 Regeneron Agreement"). The 2016 Regeneron Agreement has two
principal components: (i) a product development component under which the
parties will research, develop and commercialize CRISPR/Cas-based therapeutic
products primarily focused on genome editing in the liver; and (ii) a technology
collaboration component, pursuant to which the parties will engage in research
and development activities aimed at discovering and developing novel
technologies and improvements to CRISPR/Cas technology to enhance our genome
editing platform. We may also access the Regeneron Genetics Center and
proprietary mouse models to be provided by Regeneron for a limited number of our
liver programs. At the inception of the 2016 Regeneron Agreement, Regeneron
selected the first of its 10 targets, ATTR, which is subject to the ATTR Co/Co.

On May 30, 2020, we entered into (i) amendment no. 1 (the "2020 Regeneron
Amendment") to the 2016 Regeneron Agreement, (ii) co-development and co-funding
agreements for the treatment of hemophilia A and hemophilia B (the "Hemophilia
Co/Co") agreements and (iii) a stock purchase agreement. The collaboration
expansion builds upon the jointly developed targeted transgene insertion
capabilities designed to durably restore missing therapeutic protein, and to
overcome the limitations of traditional gene therapy. The collaboration was
extended until April 2024, at which point Regeneron has an option to renew for
an additional two years. The 2020 Regeneron Amendment also grants Regeneron
exclusive rights to develop products for five additional in vivo
CRISPR/Cas-based therapeutic liver targets and non-exclusive rights to
independently develop and commercialize up to 10 ex vivo gene edited products
made using certain defined cell types. Refer to Note 7 to our condensed
consolidated financial statements of this Quarterly Report on Form 10-Q for
additional information related to the terms of the agreement between us and
Regeneron.

AvenCell Therapeutics, Inc. ("AvenCell")



On July 30, 2021, we finalized a transaction in which we, Cellex Cell
Professionals GmbH ("Cellex") and funds managed by Blackstone Life Sciences
Advisors L.L.C. ("BXLS") established a new universal CAR-T cell therapy company,
AvenCell, and entered into two agreements with AvenCell: (i) a license and
collaboration agreement (the "LCA"), under which we will collaborate to develop
allogeneic universal CAR-T cell therapies and granted AvenCell a license to
develop and commercialize genome edited universal CAR-T cell therapies (limited
to its use with their switchable, universal CAR-T cell UniCAR and RevCAR
platforms); and (ii) a co-development and co-funding agreement (the "AvenCell
Co/Co") under which we will co-develop and co-commercialize allogeneic universal
CAR-T cell products for an immuno-oncology indication. In addition to the
license, we will collaborate with AvenCell on at least seven universal CAR-T
cell products that combine our allogeneic T cell technology with AvenCell's
switchable, universal CAR-T cell technology, referred to as the ("Allo
Collaboration"). Additionally, AvenCell will pay us to provide supply and
manufacturing services for them, including supplying good manufacturing practice
("GMP") CRISPR reagents to support the research and development of all CRISPR
Products (as defined in the LCA) under the Allo Collaboration until the
completion of the first Pivotal Trial (as defined in the LCA) of the first such
CRISPR Product. We will also have one additional option to enter into a second
co-development and co-funding agreement from selected allogeneic universal CAR-T
cell therapy products that the parties intend to develop under the Allo
Collaboration for a payment of $30.0 million to AvenCell.

In exchange for the license, we received a 33.33% equity interest in AvenCell at the time of the initial closing. Refer to Notes 7 and 8 to our condensed consolidated financial statements of this Quarterly Report on Form 10-Q for additional information related to the terms of the agreement between us and AvenCell.

SparingVision SAS ("SparingVision")



In October 2021, we and SparingVision, a genomic medicine company developing
vision saving treatments for ocular diseases, entered into a license and
collaboration agreement (the "SparingVision LCA"), to develop novel genomic
medicines utilizing CRISPR/Cas9 technology for the treatment of ocular diseases.
We will grant SparingVision exclusive rights to our proprietary in vivo
CRISPR/Cas9-based genome editing technology for up to three ocular targets
addressing diseases with significant unmet medical need. In addition, the
parties will research and develop novel self-inactivating AAV vectors and
LNP-based approaches

                                       29
--------------------------------------------------------------------------------

to address delivery of CRISPR/Cas9 genome editing reagents to the retina. SparingVision will lead and fund the preclinical and clinical development for the genome editing product candidates pursued under the collaboration.



In exchange for the license, we received an 11% equity ownership in
SparingVision as of the closing date as well as three warrants attached to each
share received for the right to purchase additional shares at designated prices
that are subject to certain vesting conditions. We will also be eligible to
receive certain research, development and commercial milestone cash payments (up
to approximately $200 million per product) as well as royalties on potential
future sales of products arising from the collaboration. We will have an option
to obtain exclusive U.S. commercialization rights for product candidates arising
from two of three collaboration targets. For product candidates we choose to
option, we will pay an opt-in fee between $10.0 million and $20.0 million
depending on the stage of development of the target, reimburse certain costs,
share in 50% of development costs and pay royalties to SparingVision on U.S.
sales. Refer to Notes 7 and 8 to our condensed consolidated financial statements
of this Quarterly Report on Form 10-Q for additional information related to the
terms of the agreement between us and SparingVision.

Kyverna Therapeutics, Inc. ("Kyverna")



In December 2021, we entered into a licensing and collaboration agreement with
Kyverna, a cell therapy company engineering a new class of therapies for
autoimmune and inflammatory diseases, for the development of an allogeneic CD19
CAR-T cell therapy for the treatment of a variety of B cell-mediated autoimmune
diseases. We granted Kyverna rights to our proprietary ex vivo CRISPR/Cas9-based
allogeneic platform for the development of KYV-201, an allogeneic CD19 CAR-T
cell investigational candidate for the treatment of select autoimmune diseases.
This is a novel approach aimed at targeting CD19 for inflammatory diseases as
compared to traditional oncology indications. Kyverna will lead and fund
preclinical and clinical development for KYV-201 and we will be eligible to
receive certain development and commercial milestone payments, as well as
low-to-mid-single-digit royalties on potential future sales. We may also
exercise an option to lead U.S. commercialization for KYV-201 under a
co-development and co-commercialization agreement. If we choose to co-develop
and co-commercialize KYV-201, we will pay an opt-in fee of $5.0 million and
share in 50% of development costs and future net profit and/or loss arising from
commercializing KYV-201 in the U.S. Kyverna retains all rights outside of the
U.S., and we will receive low-to-mid-single-digit royalties on net sales
generated outside of the U.S.

In exchange for the license, we received an equity ownership of preferred stock
in Kyverna. We separately made an additional investment in Kyverna, purchasing
incremental shares of Kyverna's preferred stock in exchange for $3.0 million in
cash, bringing our investment to approximately 7% ownership in Kyverna at the
time of closing. Refer to Notes 7 and 8 to our condensed consolidated financial
statements of this Quarterly Report on Form 10-Q for additional information
related to the terms of the agreement between us and Kyverna.

ONK Therapeutics, Ltd ("ONK")



In February 2022, we announced a license, collaboration and option agreement
with ONK for the development of engineered NK cell therapies for the treatment
of cancer. The agreement grants ONK a non-exclusive license to our proprietary
ex vivo CRISPR/Cas9-based genome editing platform and our LNP-based delivery
technologies for development of up to five allogeneic NK cell therapies. ONK
will be responsible for preclinical and clinical development for the engineered
NK cell therapies enabled by the agreement. We will be eligible to receive up to
$184 million per product in development and commercial milestone payments, as
well as up to mid-single-digit royalties on potential future sales. In addition,
the agreement grants us options to co-develop and co-commercialize up to two
products worldwide with rights to lead commercialization in the U.S. Refer to
Note 7 to our condensed consolidated financial statements of this Quarterly
Report on Form 10-Q for additional information related to the terms of the
agreement between us and ONK.

Novartis



In December 2014, we entered into the 2014 Novartis Agreement, primarily focused
on the research of new ex vivo CRISPR/Cas9-edited therapies using CAR-T cells
and HSCs. The agreement was amended in December 2018 to also include research on
OSCs. In December 2019, per the terms of the 2014 Novartis Agreement, the
research term ended, although the 2014 Novartis Agreement remains in effect, for
which we will be eligible to receive milestone and royalty payments in the
future. In June 2021, we entered into Amendment No. 3 (the "Amendment") to the
2014 Novartis Agreement. The Amendment amends Novartis' rights with respect to
all of the CAR-T Therapeutic Targets (as defined in the 2014 Novartis Agreement)
that Novartis selected under the 2014 Novartis Agreement, including (a) making
Novartis' license non-exclusive for such CAR-T Therapeutic Targets, (b) removing
Novartis' diligence and related reporting obligations for such CAR-T Therapeutic
Targets, and (c) refining the scope of Novartis' sublicense rights for such
CAR-T Therapeutic Targets. We made a one-time payment to Novartis of $10.0

                                       30
--------------------------------------------------------------------------------


million within 30 days after the effective date of the Amendment, which was
recorded as research and development expense in the consolidated statement of
operations and comprehensive loss for the year ended December 31, 2021. Since
December 31, 2021, there have been no other material changes to the key terms of
the 2014 Novartis Agreement and the Novartis Amendment. Refer to Note 7 to our
condensed consolidated financial statements of this Quarterly Report on Form
10-Q for additional information related to the terms of the agreement between us
and Novartis.

Financial Overview

Collaboration Revenue

Our revenue consists of collaboration revenue, including amounts recognized
related to upfront technology access payments for licenses, technology access
fees, research funding and milestone payments earned under our collaboration and
license agreements.

Research and Development

Research and development expenses consist of expenses incurred in performing
research and development activities, such as compensation and benefits, which
includes equity-based compensation, for full-time research and development
employees, allocated facility-related expenses, overhead expenses, license and
milestone fees, contract research, development and manufacturing services,
clinical trial costs and other related costs.

General and Administrative



General and administrative expenses consist primarily of compensation and
benefits, including equity-based compensation, for our executive, finance,
legal, human resources, business development and support functions. Also
included in general and administrative expenses are allocated facility-related
costs not otherwise included in research and development expenses, travel
expenses and professional fees for auditing, tax and legal services, including
IP-related legal services, and other consulting fees and expenses.

Other (Expense) Income, Net

Other (expense) income consists of interest income earned on our cash, cash equivalents, restricted cash equivalents and marketable securities, loss from equity method investment and change in fair value of contingent consideration.

Results of Operations

The following discussion of the financial condition and results of operations should be read in conjunction with the accompanying condensed consolidated financial statements and the related footnotes thereto.

Comparison of Three Months Ended June 30, 2022 and 2021

The following table summarizes our results of operations for the three months ended June 30, 2022 and 2021:



                                              Three Months Ended June 30,           Period-to-
                                               2022                 2021           Period Change
                                                              (In thousands)
Collaboration revenue                     $        14,030       $       6,550     $         7,480
Operating expenses:
Research and development                           90,199              58,884              31,315
General and administrative                         22,132              16,683               5,449
Total operating expenses                          112,331              75,567              36,764
Operating loss                                    (98,301 )           (69,017 )           (29,284 )
Other (expense) income, net:
Interest income                                       703                 211                 492
Loss from equity method investment                 (3,252 )                 -              (3,252 )
Change in fair value of contingent
consideration                                         172                   -                 172
Total other (expense) income, net                  (2,377 )               211              (2,588 )
Net loss                                  $      (100,678 )     $     (68,806 )   $       (31,872 )




                                       31

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

Collaboration Revenue



Collaboration revenue increased by approximately $7.5 million to $14.0 million
during the three months ended June 30, 2022, as compared to $6.6 million during
the three months ended June 30, 2021. The increase in collaboration revenue
during the three months ended June 30, 2022 is primarily due to our joint
venture with AvenCell and revenue from the Kyverna LCA. Refer to Note 7 to our
condensed consolidated financial statements appearing elsewhere in this
Quarterly Report on Form 10-Q for further details.

Research and Development



Research and development expenses increased by approximately $31.3 million to
$90.2 million during the three months ended June 30, 2022, as compared to $58.9
million during the three months ended June 30, 2021.

The following table summarizes our research and development expenses for the
three months ended June 30, 2022 and 2021, together with the changes in those
items in dollars and the respective percentages of change:

                                  Three Months Ended June 30,           Period-to-           Percent
                                   2022                 2021           Period Change          Change
                                                            (In thousands)
External development
expenses by program:
  NTLA-2001                   $        9,017       $        4,280     $         4,737                111 %
  NTLA-2002                            3,357                1,561               1,796                115 %
  NTLA-5001                            4,757                4,724                  33                  1 %
Unallocated research and
development expenses:
Employee-related expenses             28,824               15,635              13,189                 84 %
Research materials and
contracted services                   21,122               18,679               2,443                 13 %
Facility-related expenses              8,174                6,778               1,396                 21 %
Stock-based compensation              14,079                6,135               7,944                129 %
Other                                    869                1,092                (223 )              -20 %
Total research and
development expenses          $       90,199       $       58,884     $        31,315                 53 %



The increase in research and development expenses for the three months ended
June 30, 2022 compared to the three months ended June 30, 2021 was primarily
attributable to:

a $4.7 million increase in external costs related to the development of NTLA-2001, our lead product candidate, primarily due to an increase in spend on contracted services and lab consumables;


                                       32
--------------------------------------------------------------------------------

a $1.8 million increase in external costs related to the development of NTLA-2002, primarily due to an increase in spend on drug components;

a $13.2 million increase in employee-related expenses primarily driven by the expansion of our development organization;


a $2.4 million increase in research materials and contracted services primarily
driven by an increase in drug component expenses to support our pipeline,
partially offset by a $10.0 million one-time payment related to the amendment of
the 2014 Novartis Agreement in June 2021;

a $1.4 million increase in facility-related expenses primarily related to rent, depreciation and technology expense allocated to research and development; and

a $7.9 million increase in stock-based compensation driven by our larger workforce.

General and Administrative



General and administrative expenses increased by $5.4 million to $22.1 million
during the three months ended June 30, 2022, compared to $16.7 million during
the three months ended June 30, 2021. This increase was primarily related to
employee-related expenses, including an increase in stock-based compensation of
$4.5 million, driven by our larger workforce.

Other (Expense) Income, Net



The increase in other (expense) income of $2.6 million is primarily related to
our share of three months of AvenCell's losses generated in the first quarter of
2022 of $3.3 million, offset in part by a $0.5 million increase in interest
income and a reduction in the fair value of our contingent consideration
liability of $0.2 million.

Comparison of Six Months Ended June 30, 2022 and 2021

The following table summarizes our results of operations for the six months ended June 30, 2022 and 2021:



                                             Six Months Ended June 30,           Period-to-
                                              2022                2021          Period Change
                                                             (In thousands)
Collaboration revenue                     $      25,282       $     12,995     $        12,287
Operating expenses:
Research and development                        223,294             98,160             125,134
General and administrative                       44,535             30,277              14,258
Total operating expenses                        267,829            128,437             139,392
Operating loss                                 (242,547 )         (115,442 )          (127,105 )
Other (expense) income, net:
Interest income                                   1,243                431                 812
Loss from equity method investment               (5,997 )                -              (5,997 )
Change in fair value of contingent
consideration                                      (249 )                -                (249 )
Total other (expense) income, net                (5,003 )              431              (5,434 )
Net loss                                  $    (247,550 )     $   (115,011 )   $      (132,539 )


Collaboration Revenue

Collaboration revenue increased by approximately $12.3 million to $25.3 million
during the six months ended June 30, 2022, as compared to $13.0 million during
the six months ended June 30, 2021. The increase in collaboration revenue during
the six months ended June 30, 2022 is primarily due to our joint venture with
AvenCell and revenue from the Kyverna LCA. Refer to Note 7 to our condensed
consolidated financial statements appearing elsewhere in this Quarterly Report
on Form 10-Q for further details.

Research and Development



Research and development expenses increased by $125.1 million to $223.3 million
during the six months ended June 30, 2022, as compared to $98.2 million during
the six months ended June 30, 2021.

                                       33
--------------------------------------------------------------------------------


The following table summarizes our research and development expenses for the six
months ended June 30, 2022 and 2021, together with the changes in those items in
dollars (in thousands) and the respective percentages of change:


                                  Six Months Ended June 30,            Period-to-           Percent
                                   2022                2021           Period Change          Change
                                                           (In thousands)
External development
expenses by program:
  NTLA-2001                   $       17,589       $       6,602     $        10,987                166 %
  NTLA-2002                            5,057               3,716               1,341                 36 %
  NTLA-5001                            9,879               9,252                 627                  7 %
Unallocated research and
development expenses:
Employee-related expenses             53,536              29,302              24,234                 83 %
Research materials and
contracted services                   38,751              25,656              13,095                 51 %
In-process research and
development                           55,990                   -              55,990                  0 %
Facility-related expenses             16,699              11,960               4,739                 40 %
Stock-based compensation              24,353               9,626              14,727                153 %
Other                                  1,440               2,046                (606 )              -30 %
Total research and
development expenses          $      223,294       $      98,160     $       125,134                127 %

The increase in research and development expenses for the six months ended June 30, 2022 compared to the six months ended June 30, 2021 was primarily attributable to:

an $11.0 million increase in external costs related to the development of NTLA-2001, our lead product candidate, primarily due to an increase in spend on drug components, contracted services and lab consumables;

a $1.3 million increase in external costs related to the development of NTLA-2002, primarily due to an increase in spend on drug components;

a $0.6 million increase in external costs related to the development of NTLA-5001, primarily due to an increase in spend on drug components and lab consumables, offset in part by a decrease in contracted services as we entered the clinic during the first half of 2022;

a $24.2 million increase in employee-related expenses, primarily driven by the expansion of our development organization;


a $13.1 million increase in research materials and contracted services primarily
driven by an increase in drug component expenses to support our pipeline,
partially offset by a $10.0 million one-time payment related to the amendment of
the 2014 Novartis Agreement in June 2021;

$56.0 million of expense in the first half of 2022 related to the acquisition of Rewrite Therapeutics, Inc.;

a $4.7 million increase in facility-related expenses primarily related to rent, depreciation and technology expense allocated to research and development; and

a $14.7 million increase in stock-based compensation driven by our larger workforce.

General and Administrative



General and administrative expenses increased by approximately $14.3 million to
$44.5 million during the six months ended June 30, 2022, compared to $30.3
million during the six months ended June 30, 2021. This increase was primarily
related to an increase in employee related expenses, including stock-based
compensation of $9.8 million.

Other (Expense) Income, Net

                                       34

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


The increase in other (expense) income of $5.4 million is primarily related to
our share of six months of AvenCell's losses generated in the fourth quarter of
2021 and the first quarter of 2022 totaling $6.0 million and an increase in the
fair value of our contingent consideration liability of $0.2 million, offset in
part by a $0.8 million increase in interest income.

Liquidity and Capital Resources



Since our inception through June 30, 2022, we have raised an aggregate of
approximately $1,861.1 million to fund our operations through our collaboration
agreements, our initial public offering and concurrent private placements,
follow-on public offerings, at-the-market offerings and the sale of convertible
preferred stock.

As of June 30, 2022, we had $906.9 million in cash, cash equivalents and marketable securities.



We are eligible to earn a significant amount of milestone payments and
royalties, in each case, on a per-product basis under our collaborations with
Novartis, SparingVision and ONK, on a per-target basis under our collaboration
with Regeneron and upon achievement of certain events under our collaboration
with Kyverna. Our ability to earn these milestone payments and the timing of
achieving these milestones is dependent upon the outcome of our research and
development activities and is uncertain at this time. Our rights to payments
under our collaboration agreements are our only committed external source of
funds.

Follow-on Offerings

In July 2021, we closed an underwritten public offering of 4,758,620 shares of
common stock, including the exercise in full of the underwriters' option to
purchase an additional 620,689 shares of common stock, at the public offering
price of $145.00 per share, for aggregate net proceeds of $648.3 million, after
deducting approximately $41.7 million in underwriting discounts and offering
costs.

At-the-Market Offering Programs



In August 2019, we entered into an Open Market Sale Agreement (the "2019 Sale
Agreement") with Jefferies LLC ("Jefferies"), under which Jefferies is able to
offer and sell, from time to time in "at-the-market" offerings, shares of our
common stock having aggregate gross proceeds of up to $150.0 million. We agreed
to pay to Jefferies cash commissions of 3.0% of the gross proceeds of sales of
common stock under the 2019 Sale Agreement.

During 2021, we issued 641,709 shares of our common stock in a series of sales
at an average price of $72.79 per share in accordance with the 2019 Sale
Agreement, for aggregate net proceeds of $45.3 million after payment of cash
commissions to Jefferies and approximately $0.1 million related to legal,
accounting and other fees in connection with the sales.

During the first half of 2022, we issued 579,788 shares of our common stock in a
series of sales at an average price of $69.43 per share in accordance with the
2019 Sale Agreement, for aggregate net proceeds of $38.9 million after payment
of cash commissions to Jefferies and approximately $0.2 million related to
legal, accounting and other fees in connection with the sales.

As of June 30, 2022, $7.2 million in shares of our common stock remain eligible for sale under the 2019 Sale Agreement.



In March 2022, we entered into an Open Market Sale Agreement (the "2022 Sale
Agreement") with Jefferies, under which Jefferies is able to offer and sell,
from time to time in "at-the-market" offerings, shares of our common stock
having aggregate gross proceeds of up to $400.0 million. We agreed to pay to
Jefferies cash commissions of 3.0% of the gross proceeds of sales of common
stock under the 2022 Sale Agreement. No shares have been issued under the 2022
Sale Agreement through June 30, 2022.

                                       35
--------------------------------------------------------------------------------

Funding Requirements



Our primary uses of capital are, and we expect will continue to be, research and
development contracted services, clinical trial costs, compensation and related
expenses, laboratory and office facilities, research supplies, legal and
regulatory expenses, patent prosecution filing and maintenance costs for our
licensed IP, milestone and royalty payments and general overhead costs. During
the second half of 2022, we expect our expenses to increase compared to prior
periods in connection with our ongoing activities as we continue to grow our
research and development team, development of our clinical programs and advance
additional programs into clinical development.

Because our lead programs are still in the early clinical stage and the outcome
of these efforts is uncertain, we cannot estimate the actual amounts necessary
to successfully complete the development and commercialization of any future
product candidates or whether, or when, we may achieve profitability. Until such
time as we can generate substantial product revenues, if ever, we expect to
finance our ongoing cash needs through equity financings and collaboration
arrangements. We receive cost reimbursements from Regeneron for the
transthyretin ("ATTR") amyloidosis and hemophilia programs. Additionally, we are
eligible to earn milestone payments and royalties, in each case, on a
per-product basis under our collaborations with Novartis, SparingVision and ONK,
on a per-target basis under our collaboration with Regeneron, and upon
achievement of certain events with Kyverna, subject to the provisions of our
agreements with each of them. Except for these sources of funding, we will not
have any committed external source of liquidity. To the extent that we raise
additional capital through the future sale of equity, the ownership interest of
our stockholders will be diluted, and the terms of these securities may include
liquidation or other preferences that adversely affect the rights of our
existing stockholders. If we raise additional funds through collaboration
arrangements in the future, we may have to relinquish valuable rights to our
technologies, future revenue streams or product candidates or grant licenses on
terms that may not be favorable to us. If we are unable to raise additional
funds through equity financings when needed, we may be required to delay, limit,
reduce or terminate our product development or future commercialization efforts
or grant rights to develop and market product candidates that we would otherwise
prefer to develop and market ourselves.

Outlook



Based on our research and development plans and our expectations related to the
progress of our programs, we expect that our cash, cash equivalents and
marketable securities as of June 30, 2022, as well as research and cost
reimbursement funding from Regeneron, AvenCell and SparingVision, will enable us
to fund our ongoing operating expenses and capital expenditure requirements
beyond the next 24 months, excluding any potential milestone payments or
extension fees that could be earned and distributed under our collaboration
agreements or any strategic use of capital not currently in the base case
planning assumptions. We have based this estimate on current assumptions that
may prove to be wrong, and we could use our capital resources sooner than we
expect.

Our ability to generate revenue and achieve profitability depends significantly
on our success in many areas, including: developing our delivery technologies
and our CRISPR/Cas9 technology platform; selecting appropriate product
candidates to develop; completing research and preclinical and clinical
development of selected product candidates; obtaining regulatory approvals and
marketing authorizations for product candidates for which we complete clinical
trials; developing a sustainable and scalable manufacturing process for product
candidates; launching and commercializing product candidates for which we obtain
regulatory approvals and marketing authorizations, either directly or with a
collaborator or distributor; obtaining market acceptance of our product
candidates; addressing any competing technological and market developments;
negotiating favorable terms in any collaboration, licensing, or other
arrangements into which we may enter; maintaining good relationships with our
collaborators and licensors; maintaining, protecting, and expanding our
portfolio of IP rights, including patents, trade secrets, and know-how; and
attracting, hiring, and retaining qualified personnel.

Cash Flows



The following is a summary of cash flows for the six months ended June 30, 2022
and 2021:

                                                Six Months Ended June 30,
                                                2022                2021
                                                        (In millions)

Net cash used in operating activities $ (164.9 ) $ (105.4 ) Net cash provided by investing activities

           142.8                 

7.7


Net cash provided by financing activities            53.2                65.7




                                       36

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

Net cash used in operating activities



Net cash used in operating activities of $164.9 million during the six months
ended June 30, 2022 primarily reflects the increased spend in our research and
development activities, offset in part by the receipt of $3.8 million in
payments from our collaboration partners during that period. Net cash used in
operating activities of $105.4 million during the six months ended June 30, 2021
primarily reflects the increased spend in our research and development
activities, offset in part by the receipt of $3.2 million in payments from our
collaboration partners during that period.

Net cash provided by investing activities



During the six months ended June 30, 2022 and 2021, our investing activities
provided cash of $142.8 million and $7.7 million, respectively. The increase in
the six months ended June 30, 2022 is primarily due to $224.6 million in
marketable securities maturing, offset in part by $44.8 million in net cash for
the acquisition of Rewrite, $31.5 million of marketable securities purchased and
$5.4 million in cash for the purchase of property and equipment. The increase in
the six months ended June 30, 2021 is primarily due to an increase in marketable
securities activity during the period, as $185.4 million in marketable
securities were purchased and $198.5 million in marketable securities matured,
offset in part by $5.4 million for the purchase of property and equipment during
the period.

Net cash provided by financing activities



Net cash provided by financing activities of $53.2 million during the six months
ended June 30, 2022 includes $38.9 million in net proceeds from at-the-market
offerings, $13.3 million in cash received from the exercise of stock options and
$1.1 million in cash received from the issuance of shares through our employee
stock purchase plan. Net cash provided by financing activities of $65.7 million
during the six months ended June 30, 2021 includes $45.3 million in net proceeds
from at-the-market offerings, $19.5 million in cash received from the exercise
of stock options and $1.0 million in cash received from the issuance of shares
through our employee stock purchase plan.

Critical Accounting Policies



Our critical accounting policies require the most significant judgments and
estimates in the preparation of our condensed consolidated financial statements.
Management has determined that our most critical accounting policies are those
relating to revenue recognition and equity-based compensation. There have been
no changes to our critical accounting policies from those which were discussed
in our Annual Report for the year ended December 31, 2021.

Recent Accounting Pronouncements



Please read Note 2, "Summary of Significant Accounting Policies", to our
condensed consolidated financial statements included in Part I, Item 1, "Notes
to Condensed Consolidated Financial Statements," of this Quarterly Report on
Form 10-Q for a description of recent accounting pronouncements applicable to
our business.

Contractual Obligations

There were no material changes to our contractual obligations during the three
months ended June 30, 2022. For a complete discussion of our contractual
obligations, please refer to our Management's Discussion and Analysis of
Financial Condition and Results of Operations in our Annual Report for the year
ended December 31, 2021.

© Edgar Online, source Glimpses