The discussion in "Management's Discussion and Analysis of Financial Condition
and Results of Operations" contains trend analysis, estimates and other
forward-looking statements within the meaning of Section 27A of the Securities
Act of 1933, as amended, and Section 21E of the Exchange Act of 1934, as
amended. These forward-looking statements include, without limitation,
statements containing the words "anticipates," "believes," "continues," "could,"
"estimates," "expects," "intends," "may," "plans," "seeks," "should," "will,"
and other words of similar import or the negative of those terms or expressions.
Such forward-looking statements are subject to known and unknown risks,
uncertainties, estimates and other factors that may cause our actual results,
performance or achievements, or industry results, to be materially different
from any future results, performance or achievements expressed or implied by
such forward-looking statements. Actual results could differ materially from
those set forth in such forward-looking statements as a result of, but not
limited to, the "Risk Factors" described in Part I, Item 1A our Annual Report on
Form 10-K for the year ended December 31, 2021 as filed with the Securities and
Exchange Commission on February 24, 2022, or the 2021 Annual Report, as
supplemented by the risks described under "Risk Factors" in Part II, Item 1A of
this Quarterly Report on Form 10-Q. You should also read the following
discussion and analysis in conjunction with our Condensed Consolidated Financial
Statements and accompanying notes included in this report and the Consolidated
Financial Statements and accompanying notes thereto included in our 2021 Annual
Report.

Overview

We are a clinical-stage genomic medicine company committed to translating
ground-breaking science into medicines that transform the lives of patients and
families afflicted with serious diseases. We plan to deliver on this mission
through development of our clinical and preclinical product candidates
leveraging our novel science and our in-house manufacturing capabilities.

Clinical Programs Updates:



•Fabry Disease: Isaralgagene civaparvovec, also known as ST-920, is our
wholly-owned gene therapy product candidate for the treatment of Fabry disease
and is currently being evaluated in our Phase 1/2 STAAR clinical study. Sangamo
recently presented updated preliminary data from the STAAR study at three
medical conferences. Data were presented at the Society for the Study of Inborn
Errors of Metabolism (SSIEM) Annual Meeting in August, as of the
February 14, 2022 cutoff date. Updated preliminary data as of the July 21, 2022
cutoff date were presented at the 29th Congress of the European Society of Gene
& Cell Therapy, or ESGCT, and National Organization for Rare Disorders, or NORD,
conference in October. The updated preliminary data showed that all nine
patients from the dose escalation phase exhibited sustained elevated ?-Gal A
activity, ranging from nearly 2-fold to 30-fold of mean normal, for up to 23
months post dosing, as of the last date of measurement. Four patients were
withdrawn from enzyme replacement therapy, or ERT, and maintained significantly
elevated levels of ?-Gal A activity up to 28 weeks post withdrawal. Since the
July 21, 2022 cutoff date, the fifth and final patient in the dose escalation
phase who started the study on ERT was withdrawn from ERT. All patients
withdrawn have remained off ERT. The Phase 1/2 STAAR study has transitioned into
the expansion phase, with the first five expansion patients dosed at the 5e13
vg/kg dose level, including the first two female patients. We have multiple
patients in screening, including both male and female candidates. We also
continue to actively plan for a potential pivotal Phase 3 trial and are engaging
with health authorities, patient advocacy groups and investigators.

•Sickle Cell Disease: BIVV003, formerly known as SAR445136, is our zinc finger
nuclease, or ZFN, gene-edited autologous cell therapy product candidate for the
treatment of sickle cell disease, or SCD, and is currently being evaluated in
our Phase 1/2 PRECIZN-1 clinical study. In October 2022, we dosed the sixth
patient in the Phase 1/2 study, who is the second patient in the study to
receive a product candidate manufactured using improved methods that have been
shown in internal experiments to increase the number of long-term progenitor
cells in the final product. In August 2022, the FDA granted Sangamo's request
for regenerative medicine advanced therapy, or RMAT, designation for BIVV003.
RMAT designation is granted to regenerative medicine therapies intended to
treat, modify, reverse, or cure a serious condition, for which preliminary
clinical evidence indicates that the medicine has the potential to address an
unmet medical need. We plan to present updated data, of which a summary of the
accepted abstract is located below, from the Phase 1/2 PRECIZN-1 study in a
poster presentation at the 64th American Society of Hematology, or ASH, Annual
Meeting & Exposition taking place December 10-13, 2022 in New Orleans,
Louisiana. Phase 3 study design, enabling activities and manufacturing readiness
continue.

•Renal Transplant Rejection: TX200 is our wholly-owned Chimeric Antigen
Receptor, or CAR, engineered regulatory T cell, or CAR-Treg, cell therapy
product candidate for the prevention of immune-mediated rejection in HLA-A2
mismatched kidney transplantation from a living donor and is currently being
evaluated in our Phase 1/2 STEADFAST clinical study. In September 2022, we dosed
the second patient in the Phase 1/2 study. The product candidate continues
                                       27

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

Table of Contents



to be generally well tolerated in both patients. In October 2022, the second
patient in the Phase 1/2 STEADFAST study experienced a serious adverse event, or
SAE, of duodenal ileus. The patient was also noted to be COVID positive. The
Principal Investigator and the Safety Monitoring Committee for the TX200 study
assessed the SAE as unrelated to the TX200 treatment, and the patient has since
fully recovered. Clinical activities continue to progress in preparation for
patient three. We plan to provide guidance on timing of dosing of the third
patient once the kidney transplant has been scheduled and we have confirmed a
potential dosing date.

•Hemophilia A: Giroctocogene fitelparvovec, also known as SB-525, is a gene
therapy product candidate for the treatment of moderately severe to severe
hemophilia A and is the subject of our Phase 1/2 Alta study and the
registrational Phase 3 AFFINE clinical trial. We are developing giroctocogene
fitelparvovec with our collaborator Pfizer Inc., or Pfizer. We and Pfizer plan
to present updated data, of which a summary of the accepted abstract is located
below, from the Phase 1/2 Alta study in a poster presentation at ASH on December
10-13, 2022. In September 2022, we and Pfizer jointly announced that the Phase 3
AFFINE trial had re-opened recruitment following a voluntary pause and
subsequent clinical hold. Trial sites resumed enrollment in September, and
dosing is expected to resume shortly. All trial sites are anticipated to be
active by the end of 2022, and a pivotal readout is expected in the first half
of 2024.

Summary of Preliminary Safety and Efficacy Results from the Phase 1/2 PRECIZN-1 Study of BIVV003



•BIVV003 is a novel therapeutic product candidate comprising autologous CD34
hematopoietic stem progenitor cells, or HSPCs, modified ex vivo by ZFNs
targeting specifically the BCL11A gene erythroid-specific enhancer, or ESE, to
increase endogenous fetal hemoglobin, or HbF, production in erythrocytes.

•PRECIZN-1 is an ongoing first-in-human, open label, single arm, multi-site
study evaluating safety and tolerability of BIVV003 in patients (n=8; aged 18-40
years) with severe SCD across six U.S. sites.

•Eligible patients underwent mobilization and apheresis with plerixafor.
Autologous HSPC, were transfected ex vivo with ZFN messenger ribonucleic acid to
manufacture BIVV003. A single IV infusion was administered at least 72 hours
after pre-conditioning with busulfan.

•Patients were monitored for stem cell engraftment and hematopoietic recovery, adverse events, or AEs, clinical and laboratory hemolysis markers, total hemoglobin and HbF, percentage of red blood cells containing HbF, and SCD related events.

•As of the cutoff date of May 3, 2022, four patients had received BIVV003 infusions up to 125 weeks of follow-up for the longest treated patient.



•A fifth patient was dosed with BIVV003 on May 4, 2022 and has engrafted. This
patient received BIVV003 manufactured using an improved process that has been
shown in internal experiments to increase the number of ZFN-modified long-term
progenitors in the product candidate.

•Of the four patients dosed prior to the cutoff date, three have improved
clinically since BIVV003 infusion through the cutoff date. For these three
patients, the percent HbF levels stabilized at 30% or greater by 26 weeks after
BIVV003 infusion and persisted for up to 104 weeks for the longest treated
patient (2 years) (see Figure 1 below). The percentage of F cells (red blood
cells that contained HbF) were sustained at 90% or greater for up to 104 weeks.
Total hemoglobin levels were maintained in the range of 9.4-12.7 g/dL. The one
patient whose percent HbF level stabilized at less than 20% and who did not
sustain HbF levels above 10 pg per cell experienced two severe VOCs at 9- and
16-months post-infusion.
                                       28

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

Table of Contents

Figure 1: Total Hb and Hb fractionation over time



[[Image Removed: sgmo-20220930_g1.jpg]]
[[Image Removed: sgmo-20220930_g2.jpg]]
                                       29
--------------------------------------------------------------------------------
  Table of     Contents
[[Image Removed: sgmo-20220930_g3.jpg]]
[[Image Removed: sgmo-20220930_g4.jpg]]

As of the May 3, 2022 cutoff date, BIVV003 was generally well tolerated with no infusion related reactions. AEs reported were consistent with plerixafor mobilization and busulfan myeloablative conditioning. No AEs or SAEs were reported that were assessed as related to BIVV003.


                                       30

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

Table of Contents

Summary of Updated Preliminary Results from the Phase 1/2 Alta Study of Giroctocogene Fitelparvovec



Eleven male patients participated in the study overall, with five patients in
the 3e13-vg/kg highest dose cohort. As of the May 20, 2022 cutoff date, patients
had been followed for 147 to 247 weeks, with two not having completed three
years (156 weeks).

•As of the May 20, 2022 cutoff date, the most commonly reported
treatment-related AEs in the highest dose cohort (3e13-vg/kg) included elevated
liver enzymes and infusion-related reactions: increased alanine
aminotransferase, or ALT 3/5 (60.0%), increased aspartate aminotransferase, or
AST 2/5 (40.0%), pyrexia 3/5 (60.0%), and tachycardia 2/5 (40.0%).

•Treatment-related SAEs were reported in one patient in the highest dose cohort
who experienced hypotension and fever with onset approximately six hours after
giroctocogene fitelparvovec infusion; the events fully resolved with treatment
and did not delay post-infusion discharge the next day. ALT elevations requiring
more than seven days of corticosteroid treatment were observed in four of the
five patients in the highest dose cohort as of the May 20, 2022 cutoff date;
elevations in ALT were managed with a tapering course of corticosteroids (median
56 days; range: 7-135 days), with maintenance of clinically meaningful levels of
factor VIII, or FVIII, activity, as evidenced by a lack of bleeding events
around the time of corticosteroid treatment and minimal bleeding events
afterwards.

•As of the May 20, 2022 cutoff date, no patient in the study developed an inhibitor to FVIII, and there have been no thrombotic events and no hepatic masses detected.



•The three patients in the 3e13-vg/kg cohort with available data through Week
156 had mean FVIII activity maintained in the mild to normal range, as shown in
the table below. Of the data available for the remaining two patients, one
maintained FVIII activity levels in the mild range through Week 130 and one had
FVIII activity levels below lower level of quantification (<3%) as measured with
a chromogenic assay and 5.4% measured with a 1-stage assay at Week 130.

•In the 3e13-vg/kg cohort, the annualized bleeding rate, meaning the number of
all bleeding episodes starting three weeks after the product candidate infusion
divided by the observation period in years, was zero for the first year
post-infusion and 0.9 throughout the total duration of follow-up. In this
cohort, two patients experienced a total of 12 bleeding events (six traumatic,
four spontaneous, two unknown) necessitating treatment with exogenous FVIII. As
of the May 20, 2022 cutoff date, no patients in this cohort have resumed
prophylaxis.

Table. Factor VIII Activity Levels by 1-Stage and Chromogenic Assay for the Giroctocogene Fitelparvovec 3e13-vg/kg Cohort



Factor VIII Activity,                                                              Study Week
% Normal, Median (min, max)
Assay                             Week 12          Week 24          Week 52         Week 78        Week 104         Week 130          Week 156
1-stage clotting                    93.7            104.8             31.1           57.5            27.5             23.3              22.9
                               (82.7, 167.7)    (30.5, 212.6)    (12.0, 191.3)   (3.8, 144.2)    (4.1, 99.1)      (5.4, 164.5)      (22.6, 129.0)
Chromogenic                         62.1             70.1             20.1           40.1            16.3             12.3              12.5
                               (51.8, 109.5)    (20.4, 123.8)     (7.8, 122.3)   (0.9, 114.7)    (0.9, 71.6)      (0.9, 113.2)      (11.8, 91.1)
Patients, n                          5                5                4a             4a              5                4a                3b

(a) There was one patient each who was unable to attend visits at Weeks 52, 78, and 130. (b) Two patients had not yet reached Week 156 at the time of the data cut. min, max = minimum, maximum

Preclinical Programs



Our preclinical development is focused in two innovative priority areas: (i)
CAR-Treg cell therapies for autoimmune disorders and (ii) genome engineering for
neurological diseases. Indications for our preclinical programs include
neurodevelopmental disorders, cancer, inflammatory bowel disease, tauopathies
and neurodegenerative diseases such as amyotrophic lateral sclerosis, multiple
sclerosis, and Huntington's disease, some of which we are developing with our
collaborators Biogen MA, Inc. and Biogen International GmbH, which we refer to
together as Biogen, Novartis Institutes for BioMedical Research, Inc., or
Novartis, Pfizer, and Takeda Pharmaceutical Company Limited.

Collaborations



Our multiple collaborations with biopharmaceutical companies bring us important
financial and strategic benefits and reinforce the potential of our research and
development efforts and our ZFP technology platform. They leverage our
collaborators' therapeutic and clinical expertise and commercial resources with
the goal to bring our medicines more rapidly to
                                       31

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

Table of Contents



patients. We believe these collaborations reflect the value of our ZFP
technology platform and will potentially expand the addressable markets of our
product candidates. To date, we have received approximately $815.0 million in
upfront licensing fees, milestone payments and proceeds from sales of our common
stock to collaborators and have the right to earn up to $6.7 billion in future
milestone payments from our collaborations, in addition to potential product
royalties.

In-House Manufacturing

We believe that our in-house manufacturing capacity provides us a competitive
advantage. We currently operate an adeno-associated virus, or AAV, manufacturing
facility in our Brisbane, California headquarters and cell therapy manufacturing
facilities in Brisbane, California and Valbonne, France. Our manufacturing
strategy is to provide greater flexibility, quality and control to our product
candidate pipeline by building a balanced and necessary capacity achieved
through our in-house manufacturing, contract manufacturing organizations, or
CMOs, and partnerships; investing in manufacturing processes and analytics; and
developing a strong supply chain.

For additional information regarding our business, see "Business" in Part I, Item 1 of the 2021 Annual Report.

Impacts of the Ongoing COVID-19 Pandemic



We have experienced and continue to experience impacts from the ongoing and
evolving COVID-19 pandemic on our business and operations and could continue to
experience these or potentially more severe impacts as the pandemic evolves in
the United States, France, the United Kingdom and locations of our clinical
studies and trials, such as the new sites for our STAAR study in Canada, Italy
and Australia. For example, we have experienced periodic short-term disruptions
to our onsite operations while addressing positive cases of COVID-19 by onsite
workers and clinical trial patients, and our operations could experience longer
term disruptions in the future in the event of a significant outbreak of
COVID-19 among our onsite workers or clinical trial patients. Moreover, from
time to time, we have been required to reorganize and prioritize our resources
to mitigate moderate COVID-19 impacts arising from travel restrictions, density
restrictions and supply constraints. If our programs encounter longer-term
disruptions, it could impact our ability to support our biopharmaceutical
partners as contemplated in our collaboration agreements and could result in
adjustments to our timelines.

Additionally, our Phase 1/2 STAAR clinical study evaluating isaralgagene
civaparvovec has experienced and continues to experience delays in its timeline
due in part to COVID-19 impacts and the diversion of healthcare resources to
fight the pandemic. For example, we have experienced delays in recruiting,
enrolling and dosing patients for this study, due in part to the hesitation of
patients to travel by plane to trial sites not within driving distance and to
enter medical facilities during the pandemic and also due in part to trial sites
prioritizing COVID-19 clinical care over research activities such as the STAAR
study. The study has also experienced delays when certain patients have decided
to take the COVID-19 vaccine or tested positive for COVID-19 prior to enrollment
or dosing in the study, and when certain patient candidates decided not to take
the COVID-19 vaccine, which disqualified them from study participation.
Moreover, we had experienced some short-term delays in sourcing the necessary
raw materials to manufacture supplies for the STAAR study and in transporting
clinical trial materials due to COVID-19 impacts. We estimated that these
challenges set back our initial STAAR study timelines by approximately three to
six months. Clinical timelines for this study could be revised again if COVID-19
impacts to our recruitment, screening, enrollment and dosing of patients and to
our sourcing of raw materials for this study intensify because of vaccination
delays, new COVID-19 variants or unexpected events.

In addition, our STEADFAST study evaluating TX200, our wholly-owned CAR-Treg
cell therapy product candidate for the treatment of kidney transplant rejection,
has experienced delays in its timeline due to COVID-19 impacts related to
manufacturing and technology transfer challenges with our CMOs and due to
patients and donors testing positive for COVID-19. Our timelines for this study
could be adjusted if COVID-19 impacts result in additional delays.

With respect to our partnered programs, the timelines for the studies and trials
managed by our collaborators are also subject to potential delay in the future
if these studies and trials experience similar challenges that we have
experienced and continue to experience in our STAAR and STEADFAST studies.

Going forward, we will continue to monitor the impact of COVID-19 on our
operations, research commitments and clinical trials and those of our
collaborators, clinical trial sites and CMOs. The magnitude of these impacts
will depend, in part, on the length and severity of the COVID-19 pandemic and
related government orders and restrictions, and how the pandemic limits the
ability of us and our business partners to operate business in the ordinary
course. Disruptions to these operations, and possibly more severe disruptions in
the future that could arise due to restrictions applicable in the places we
operate or our industry generally or to us and our facilities specifically,
could impede our ability to conduct research in a timely manner, comply with our
research obligations to our collaborators and advance the development of our
therapeutic programs. These delays and disruptions could result in adverse
material impacts to our business, operating results and financial condition.

We do not anticipate any material negative impact on our financial condition in
2022 as a result of the COVID-19 pandemic. We believe we are well positioned
financially in the near term to execute on our wholly-owned and partnered
research
                                       32

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

Table of Contents



and clinical programs. As of September 30, 2022, we had $350.3 million in cash,
cash equivalents, and marketable securities. Although we believe we are
well-capitalized currently, the effects of the evolving pandemic (along with the
effects of war in Ukraine, inflation, climate change, rising interest rates and
other economic uncertainty and volatility) could result in further disruption of
global financial markets, impairing our ability to access capital, which could
negatively affect our liquidity in the future. We do not currently anticipate
any material impairments to the valuation of the financial assets or goodwill on
our balance sheet as a result of the COVID-19 pandemic. We do not believe that
the remote workplace arrangements we have implemented for our office-based
employees have affected our financial reporting or control systems.

The extent to which the COVID-19 pandemic will impact our business, operations
and financial condition, either directly or indirectly, will depend on future
developments that remain highly uncertain at the present time. These
developments include the ultimate duration and severity of the pandemic, the
impacts of new COVID-19 variants, travel restrictions, new public health
restrictions in the United States, France, the United Kingdom and other
countries, business closures or business disruptions and the effectiveness and
timeliness of actions taken in the United States, France, the United Kingdom and
other countries to contain and treat the disease, including the effectiveness
and timing of vaccination programs. The surge of new variants of the virus,
including the recent Omicron variant and its subvariants, has resulted and may
in the future result in the return of prior, or imposition of new, orders and
restrictions. As our understanding of events evolves and additional information
becomes available, we may materially change our guidance relating to our
revenues, expenses and timelines for manufacturing, clinical trials and research
and development.

See the section titled "Risk Factors" included in Part I, Item 1A of the 2021
Annual Report for additional information on risks and uncertainties related to
the evolving COVID-19 pandemic.

Certain Components of Results of Operations



Our revenues have consisted primarily of revenues from upfront licensing fees,
reimbursements for research services, milestone achievements and research grant
funding. We expect revenues to continue to fluctuate from period to period and
there can be no assurance that new collaborations or partner reimbursements will
continue beyond their initial terms or that we are able to meet the milestones
specified in these agreements.

We have incurred net losses since inception and expect to incur losses for at least the next several years as we continue our research and development activities. To date, we have funded our operations primarily through the issuance of equity securities and revenues from collaborations and research grants.



We expect to continue to devote substantial resources to research and
development in the future and expect research and development expenses to
increase in the next several years if we are successful in advancing our product
candidates from research stage through clinical trials. Pursuant to the terms of
our agreements with Biogen, Kite Pharma, Inc., or Kite, and Novartis, certain
expenses related to research and development activities will be reimbursed to
us. The reimbursement funds to be received from Biogen, Kite, and Novartis will
be recognized as revenue as the related costs are incurred and collection is
reasonably assured.

General and administrative expenses consist primarily of salaries and personnel
related expenses for executive, finance and administrative personnel,
stock-based compensation expenses, professional fees, allocated facilities
expenses, patent prosecution expenses and other general corporate expenses. As
we continue to advance our product candidates into and through the clinic, we
expect the growth of our business to require increased general and
administrative expenses.

Critical Accounting Policies and Estimates



The accompanying management's discussion and analysis of our financial condition
and results of operations are based upon our Condensed Consolidated Financial
Statements and the related disclosures, which have been prepared in accordance
with generally accepted accounting principles in the United States. The
preparation of these Condensed Consolidated Financial Statements requires us to
make estimates, assumptions and judgments that affect the reported amounts in
our Condensed Consolidated Financial Statements and accompanying notes. We base
our estimates on historical experience and on various other assumptions that we
believe to be reasonable under the circumstances, the results of which form the
basis for making judgments about the carrying values of assets and liabilities
that are not readily apparent from other sources. Actual results may differ from
these estimates under different assumptions or conditions.

We believe our critical accounting policies and estimates relating to revenue
recognition and valuation of long-lived assets including goodwill and intangible
assets are the most significant estimates and assumptions used in the
preparation of our Condensed Consolidated Financial Statements.

There have been no significant changes in our critical accounting policies and
estimates during the three and nine months ended September 30, 2022, as compared
to the critical accounting policies and estimates disclosed in "Management's
Discussion and Analysis of Financial Condition and Results of Operations"
included in Part II, Item 7 of the 2021 Annual Report.
                                       33

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

Table of Contents



Results of Operations for the Three and Nine Months Ended September 30, 2022 and
2021

Revenues
                                            Three Months Ended September 30,                                             Nine Months Ended September 30,
                                        (in thousands, except percentage values)                                     (in thousands, except percentage values)
                                2022                 2021             Change              %                   2022                 2021             Change            %
Revenues                 $        26,460          $ 28,563          $ (2,103)            (7%)          $        84,069          $ 82,715          $ 1,354             2%


Total revenues primarily consisted of revenues from collaboration agreements. We
anticipate revenues over the next several years will be derived primarily from
our collaboration agreements with Biogen, Novartis, Kite, and Pfizer as we
continue to recognize upfront and milestone payments received under such
agreements over time.

The decrease of $2.1 million in revenues for the three months ended
September 30, 2022, compared to the same period in 2021, was primarily
attributed to a decrease of $1.9 million in revenue related to our collaboration
agreement with Novartis, and a decrease of $1.6 million in revenue related to
our collaboration agreement with Biogen. These decreases were partially offset
by a $1.9 million adjustment to revenue related to the collaboration agreement
with Sanofi S.A., or Sanofi, during 2021, primarily due to a change in estimate
regarding project costs, resulting in an adjustment to revenue under the
agreement, offset by a decrease of $1.1 million in revenue due to the
termination of collaboration agreement with Sanofi, and an increase of
$0.5 million in revenue related to our collaboration agreement with Kite.

The increase of $1.4 million in revenues for the nine months ended September 30,
2022, compared to the same period in 2021, was primarily attributed to a $1.9
million adjustment to revenue related to the collaboration agreement with Sanofi
during 2021, primarily due to a change in estimate regarding project costs,
resulting in an adjustment to revenue under the agreement, offset by a decrease
of $0.1 million in revenue due to the termination of collaboration agreement
with Sanofi, an increase of $0.5 million in revenue related to our collaboration
agreement with Kite, and an increase of $0.5 million in revenue related to our
collaboration agreement with Novartis. These increases were partially offset by
a decrease of $1.5 million in revenue related to our collaboration agreement
with Biogen.

Operating expenses
                                                      Three Months Ended September 30,                                                Nine Months Ended September 30,
                                                  (in thousands, except percentage values)                                        (in thousands, except percentage values)
                                           2022                 2021             Change             %                  2022                       2021             Change             %
Operating expenses:
Research and development            $        65,116          $ 62,498          $ 2,618             4%           $       183,719               $ 179,018          $ 4,701              3%
General and administrative                   16,238            14,501            1,737             12%                   46,239                  47,135             (896)            (2%)
Total operating expenses            $        81,354          $ 76,999          $ 4,355             6%           $       229,958               $ 226,153          $ 3,805              2%

Research and Development Expenses



Research and development expenses consisted primarily of compensation related
expenses including stock-based compensation, laboratory supplies, preclinical
and clinical studies costs, clinical supply manufacturing costs, contracted
research, and facilities and information technology expenses.

The increase of $2.6 million in research and development expenses for the three
months ended September 30, 2022, compared to the same period in 2021, was
primarily attributable to higher facilities and information technology costs of
$2.7 million driven by overall cost increases and progress made on projects
resulting in reassignment of additional space to our research and development
departments, an increase of $0.8 million in compensation and other personnel
costs as a result of increased headcount to support our programs, and an
increase of $0.5 million in travel and entertainment expenses. These increases
were partially offset by a decrease of $0.4 million in preclinical, clinical and
lab supply expenses due to the timing of certain research and development
activities, and $0.9 million in reimbursement of certain research and
development expenses by Sanofi. Stock-based compensation expense included in
research and development expenses was $4.4 million and $4.9 million for the
three months ended September 30, 2022 and 2021, respectively.

The increase of $4.7 million in research and development expenses for the nine
months ended September 30, 2022, compared to the same period in 2021, was
primarily driven by higher facilities and information technology costs of $7.8
million driven by overall cost increases and progress made on projects resulting
in reassignment of additional space to our research and development departments,
an increase of $0.9 million in travel and entertainment expenses, and an
increase of $0.8 million in compensation and other personnel costs as a result
of increased headcount to support our programs. These increases were partially
offset by a decrease of $3.7 million in preclinical, clinical and lab supply
expenses due to the timing of certain research and development activities, and
$0.9 million in reimbursement of certain research and development expenses by
Sanofi. Stock-based
                                       34

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

Table of Contents

compensation expense included in research and development expenses was $13.7 million and $14.6 million for the nine months ended September 30, 2022 and 2021, respectively.

We expect to continue to devote substantial resources to research and development in the future and expect research and development expenses to increase in the next several years if we are successful in advancing our clinical programs and if we are able to progress our earlier stage product candidates into clinical trials.



The length of time required to complete our development programs and our
development costs for those programs may be impacted by the scope and timing of
enrollment in clinical trials for our product candidates, our decisions to
pursue development programs in other therapeutic areas, and whether we pursue
development of our product candidates with a partner or collaborator or
independently. For example, our product candidates are being developed in
multiple therapeutic areas, and we do not yet know how many of those therapeutic
areas we will continue to pursue. Furthermore, the scope and number of clinical
trials required to obtain regulatory approval for each pursued therapeutic area
is subject to the input of the applicable regulatory authorities, and we have
not yet sought such input for all potential therapeutic areas that we may elect
to pursue, and even after having given such input, applicable regulatory
authorities may subsequently require additional clinical studies prior to
granting regulatory approval based on new data generated by us or other
companies, or for other reasons outside of our control. As a condition to any
regulatory approval, we may also be subject to post-marketing development
commitments, including additional clinical trial requirements. As a result of
the uncertainties discussed above, we are unable to determine the duration of or
complete costs associated with our development programs.

Our potential therapeutic products are subject to a lengthy and uncertain
regulatory process that may not result in our receipt of any necessary
regulatory approvals. Failure to receive the necessary regulatory approvals
would prevent us from commercializing the product candidates affected. In
addition, clinical trials of our product candidates may fail to demonstrate
safety and efficacy, which could prevent or significantly delay regulatory
approval. A discussion of the risks and uncertainties with respect to our
research and development activities, including completing the development of our
product candidates, and the consequences to our business, financial position and
growth prospects can be found in "Risk Factors" in Part I, Item 1A of the 2021
Annual Report, as supplemented by the risks described under "Risk Factors" in
Part II, Item 1A of this Quarterly Report on Form 10-Q.

General and Administrative Expenses



General and administrative expenses consist primarily of compensation related
expenses including stock-based compensation for executive, legal, finance and
administrative personnel, professional fees, facilities and information
technology expenses, and other general corporate expenses.

The increase of $1.7 million in general and administrative expenses for the
three months ended September 30, 2022, compared to the same period in 2021, was
primarily driven by an increase of $2.1 million in compensation and other
personnel costs, and an increase of $1.0 million in legal and professional fees.
These increases were partially offset by a decrease of $1.6 million in allocated
costs attributable to progress made on projects resulting in reassignment of
additional space to our research and development departments. Stock-based
compensation expense included in general and administrative expenses was
$3.4 million and $2.9 million for the three months ended September 30, 2022 and
2021, respectively.

The decrease of $0.9 million in general and administrative expenses for the nine
months ended September 30, 2022, compared to the same period in 2021, was
primarily driven by a decrease of $4.3 million in allocated costs attributable
to progress made on projects resulting in reassignment of additional space to
our research and development departments. This decrease was partially offset by
an increase of $3.4 million in compensation and other personnel costs, legal and
professional fees, and travel and entertainment expenses. Stock-based
compensation expense included in general and administrative expenses was
$9.7 million and $10.2 million for the nine months ended September 30, 2022 and
2021, respectively.

As we continue to build out our product portfolio and advance our product candidates into the clinic, we expect higher general and administrative expenses to support the growth of the business.

Interest and other income, net



Interest and other income, net, increased by $0.9 million for the three months
ended September 30, 2022, compared to the same period in 2021, primarily driven
by an increase of $1.2 million in interest income reflecting increases in market
interest rates, and a decrease in interest expense of $0.2 million related to
dissolution of the repayment obligation of a grant from California Institute for
Regenerative Medicine associated with the discontinuation of the ST-400 program
in 2021. These increases were partially offset by a decrease of $0.5 million
related to fluctuations in foreign currency exchange rates.

Interest and other income, net, increased by $2.7 million for the nine months
ended September 30, 2022, compared to the same period in 2021, as we benefited
from $3.0 million of Employee Retention Credit under the Coronavirus Aid,
Relief, and Economic Security Act, an increase of $1.4 million in interest
income reflecting increase in market interest rates, and a decrease
                                       35

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

Table of Contents

in interest expense of $0.6 million related to dissolution of the repayment obligation of a grant from California Institute for Regenerative Medicine associated with the discontinuation of the ST-400 program in 2021. These increases were partially offset by a decrease of $2.4 million related to fluctuations in foreign currency exchange rates.

Liquidity and Capital Resources

Liquidity



Since inception, we have incurred significant net losses, and we have funded our
operations primarily through the issuance of equity securities, payments from
corporate collaborators and strategic partners and research grants.

As of September 30, 2022, we had cash, cash equivalents, and marketable
securities totaling $350.3 million, compared to $464.7 million as of
December 31, 2021. Our most significant use of capital was for employee
compensation and external research and development expenses, including
manufacturing, clinical trials and preclinical activity related to our
therapeutic programs. Our cash and investment balances are held in a variety of
interest-bearing instruments, including U.S. government-sponsored entity debt
securities, commercial paper securities, money market funds, corporate debt
securities, asset-backed securities and certificates of deposit. Cash in excess
of immediate requirements is invested in accordance with our investment policy
with a view toward capital preservation and liquidity.

In August 2020, we entered into an Open Market Sale Agreement?, or the sales
agreement, with Jefferies LLC, providing for the sale of up to $150.0 million of
our common stock from time to time in "at-the-market" offerings under an
existing shelf registration statement. During the nine months ended
September 30, 2022, we sold 14,711,770 shares of our common stock under the
sales agreement for net proceeds of approximately $66.4 million, of which
approximately $0.6 million was received in October 2022 and recorded within
prepaid expenses and other current assets on our Condensed Consolidated Balance
Sheet as of September 30, 2022. Subsequently in October, we sold 1,921,594
shares of our common stock under the sales agreement for net proceeds of
approximately $8.7 million.

While we expect our rate of cash usage to increase in the future, in particular
to support our product development endeavors, we currently believe that our
available cash, cash equivalents, and marketable securities and expected
revenues from collaborations and strategic partnerships will be adequate to fund
our currently planned operations through at least the next 12 months from the
date our Condensed Consolidated Financial Statements are issued. We expect to
raise additional capital through additional collaborative agreements and/or the
sale of additional equity or debt financing to fund our future needs beyond the
next 12 months. During this period of uncertainty and volatility, we will
continue to monitor our liquidity.

Cash Flows

Operating activities



Net cash used in operating activities was $166.6 million for the nine months
ended September 30, 2022, primarily reflecting our net loss of $140.3 million, a
decrease in deferred revenues of $65.0 million, an increase in prepaid expenses
and other assets by $5.8 million, and a decrease in lease liabilities by
$3.3 million. These decreases were partially offset by $38.2 million of non-cash
expenses related to stock-based compensation, depreciation and amortization,
amortization of premium on marketable securities, and amortization of operating
lease right-of-use assets and a $9.4 million increase in accounts payable and
other accrued liabilities.

Net cash used in operating activities was $180.5 million for the nine months
ended September 30, 2021, primarily reflecting our net loss of $140.8 million, a
decrease in deferred revenues of $62.3 million, an increase in prepaid expenses
and other assets by $6.5 million, a decrease in accounts payable and other
accrued liabilities by $4.7 million, a decrease in long term portion of lease
liabilities by $3.2 million, an increase in accounts receivable by $2.7 million,
and a decrease in accrued compensation and employee benefits by $1.9 million.
These decreases were partially offset by $40.1 million of non-cash expenses
related to stock-based compensation, depreciation and amortization, amortization
of premium (discount) on marketable securities, and amortization and other
changes in operating lease right-of-use assets.

Investing activities

Net cash provided by investing activities was $16.7 million for the nine months ended September 30, 2022, related to maturities of marketable securities of $255.0 million, partially offset by purchases of marketable securities of $225.6 million, and $12.7 million from purchases of property and equipment.

Net cash provided by investing activities for the nine months ended September 30, 2021 was $195.6 million, mostly related to maturities of marketable securities of $509.6 million and sale of marketable securities of $6.9 million, partially offset by purchases of marketable securities of $300.4 million, and purchases of property and equipment of $20.4 million.


                                       36

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

Table of Contents

Financing activities



Net cash provided by financing activities was $65.1 million for the nine months
ended September 30, 2022, mostly related to $67.5 million of proceeds from the
at-the-market offering, net of offering expenses of $1.7 million, and proceeds
from purchases of common stock under the employee stock purchase plan of
$1.1 million, partially offset by taxes paid related to net share settlement of
equity awards of $2.0 million.

Net cash provided by financing activities was $30.7 million for the nine months
ended September 30, 2021, mostly related to $27.9 million of proceeds from the
at-the-market offering, net of offering expenses of $0.8 million, and proceeds
from the exercise of stock options and purchases under the employee stock
purchase plan of $6.6 million, partially offset by taxes paid related to net
share settlement of equity awards of $3.0 million.

Operating Capital and Capital Expenditure Requirements



We anticipate continuing to incur operating losses for at least the next several
years. Although we believe we are well capitalized currently, the effects of the
ongoing COVID-19 pandemic, war in Ukraine, inflation, rising interest rates and
other economic uncertainty and volatility could result in significant disruption
of global financial markets, impairing our ability to access capital, which
could in the future negatively affect our liquidity. Future capital requirements
beyond the next 12 months will be substantial, and we will need to raise
substantial additional capital to fund the development, manufacturing and
potential commercialization of our product candidates through additional
collaboration agreements and/or equity or debt financing. In addition, as we
focus our efforts on proprietary human therapeutics, we will need to seek FDA
approvals of our product candidates, a process that could cost in excess of
hundreds of millions of dollars per product. We regularly consider fund-raising
opportunities and may continue to decide, from time to time, to raise additional
capital based on various factors, including market conditions and our plans of
operation. Additional capital may not be available on terms acceptable to us, or
at all. If adequate funds are not available, or if the terms of potential
funding sources are unfavorable, our business and our ability to advance our
product candidate pipeline would be harmed. Furthermore, any sales of additional
equity securities, including additional sales pursuant to our at-the-market
offering program, may result in dilution to our stockholders, and any debt
financing may include covenants that restrict our business.

Our future capital requirements will depend on many forward-looking factors, including the following:

•the initiation, progress, timing and completion of clinical trials for our product candidates and potential product candidates;

•the outcome, timing and cost of regulatory approvals;

•the success of our collaboration agreements;

•delays that may be caused by changing regulatory requirements;

•the number of product candidates that we pursue;

•the costs involved in filing and prosecuting patent applications and enforcing and defending patent claims;

•the timing and terms of future in-licensing and out-licensing transactions;

•the cost and timing of establishing sales, marketing, manufacturing and distribution capabilities;

•the cost of procuring clinical and commercial supplies of our product candidates;

•the extent to which we acquire or invest in businesses, products or technologies, including the costs associated with such acquisitions and investments; and

•the costs of potential disputes and litigation.

Contractual Obligations



Our future minimum contractual obligations as of December 31, 2021 were reported
in the 2021 Annual Report. During the nine months ended September 30, 2022,
there have been no other material changes outside the ordinary course of our
business from the contractual obligations previously disclosed in our 2021
Annual Report.

© Edgar Online, source Glimpses