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 Securities Exchange Act of 1934,
as amended. These forward-looking statements reflect our current views with
respect to future events, are based on assumptions and involve known and unknown
risks, uncertainties and other factors that may cause our actual results,
performance or achievements to be materially different from any future results,
performances or achievements expressed or implied by the forward-looking
statements. When reviewing the discussion below, you should keep in mind the
substantial risks and uncertainties that could impact our business. In
particular, we encourage you to review the risks and uncertainties summarized
under "Special Note Regarding Forward-Looking Statements" that appears in the
forepart of this report and as discussed in more detail under "Risk Factors" in
Part I, Item 1A of our Annual Report on Form 10-K for the year ended
December 31, 2020 as filed with the Securities and Exchange Commission on
February 24, 2021, or the 2020 Annual Report, as supplemented by the risks and
uncertainties described under "Risk Factors" in Part II, Item 1A of this
Quarterly Report. 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 2020 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 with
serious diseases. We plan to deliver on this mission through development of our
clinical and preclinical product candidates based on our novel science and our
in-house manufacturing capabilities.
Our current clinical-stage product candidates are:
•Giroctocogene fitelparvovec, also known as SB-525, our lead product candidate,
is a gene therapy for the treatment of severe hemophilia A and is the subject of
the registrational Phase 3 AFFINE clinical trial. We are developing
giroctocogene fitelparvovec with our collaborator Pfizer Inc., or Pfizer;
•Isaralgagene civaparvovec, also known as ST-920, our wholly-owned gene therapy
product candidate for the treatment of Fabry disease, is currently being
evaluated in our Phase 1/2 STAAR clinical study and we have initiated plans for
a Phase 3 clinical trial;
•SAR445136, our zinc finger nuclease, or ZFN, gene-edited cell therapy product
candidate for the treatment of sickle cell disease, is currently being evaluated
in our Phase 1/2 PRECIZN-1 clinical study. We are developing SAR445136 with our
collaborator Sanofi S.A., or Sanofi; and
•TX200, our wholly-owned Chimeric Antigen Receptor, or CAR, engineered
regulatory T cell, or CAR-Treg, cell therapy product candidate for the treatment
of HLA-A2 mismatched kidney transplant rejection, is currently being evaluated
in our Phase 1/2 STEADFAST clinical study.
Moreover, we are focusing our preclinical development in two priority areas:
(i) CAR-Treg cell therapies for autoimmune disorders such as inflammatory bowel
disease and multiple sclerosis, and (ii) genome engineering therapies for
central nervous system, or CNS, diseases such as Alzheimer's, autism spectrum
disorder and amyotrophic lateral sclerosis, 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, and Pfizer.
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 zinc finger protein, or ZFP, technology platform.
They leverage our collaborators' therapeutic and clinical expertise and
commercial resources with the goal to bring our medicines more rapidly to
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 the sale of our
common stock to collaborators, and we are eligible to earn up to $6.9 billion in
future milestone payments from our collaborations, in addition to potential
product royalties.
We believe that our current and future in-house manufacturing capacity provides
us a competitive advantage. We currently operate an in-house adeno-associated
virus, or AAV, manufacturing facility in our Brisbane, California headquarters,
and we recently completed and brought online a cell therapy manufacturing
facility in Brisbane, California. We are also building another cell therapy
manufacturing facility in Valbonne, France, which we expect to be operational by
the end of 2021. We believe our manufacturing strategy, which leverages in-house
manufacturing and the resources of our contracting manufacturing organizations,
or CMOs, provides us flexibility, quality, control and the necessary capacity.
                                       31
--------------------------------------------------------------------------------
  Table of     Contents
Business Updates
•We announced preliminary clinical data from the first four patients treated in
our Phase 1/2 STAAR study evaluating isaralgagene civaparvovec for the treatment
of Fabry disease. The data, a summary of which is located below, showed that, as
of the September 17, 2021 cutoff date, isaralgagene civaparvovec was generally
well tolerated in the four patients treated in the first two dose cohorts. All
four patients exhibited above normal ?-Gal A activity through 14 weeks for the
most recently treated patient and one year for the first treated patient. Plasma
Lyso-Gb3 levels decreased by approximately 40% in the one patient with elevated
levels pre-treatment. In addition, we recently dosed the fifth patient in the
study, who is the first patient in the third dose cohort. We are currently
screening the sixth patient in the study. We expect to present updated clinical
data from this study throughout 2022 and present data at a medical meeting.
Based on the preliminary clinical data, we have initiated planning for a Phase 3
clinical trial of isaralgagene civaparvovec.
•We announced preliminary proof-of-concept clinical data from our Phase 1/2
PRECIZN-1 study of SAR445136 for the treatment of sickle cell disease, or SCD,
that we are developing with Sanofi. The data, a summary of which is located
below, showed that, as of the June 25, 2021 cutoff date, none of the four
treated patients required blood transfusions post-engraftment or experienced
adverse or serious adverse events related to treatment through 13 weeks of
follow-up for the most recently treated patient and 65 weeks of follow-up for
the first treated patient. The four treated patients all experienced increases
in total hemoglobin, fetal hemoglobin and percent F cells. We and Sanofi will be
presenting additional clinical data from this study at the 63rd Annual Meeting
of the American Society of Hematology, or ASH, on December 12, 2021. Based on
the preliminary proof-of-concept clinical data, we and Sanofi continue to
advance the program forward. We recently obtained manufacturing requirements
guidance from the U.S. Food and Drug Administration, or FDA, in preparation for
further potential clinical studies.
•In order to prioritize the development of SAR445136, we and Sanofi have made
the business decision to discontinue the development of ST­400, our cell therapy
product candidate for the treatment of transfusion dependent beta thalassemia.
ST-400 was developed with the support of a grant from the California Institute
for Regenerative Medicine, or CIRM.
•We announced updated clinical data from our Phase 1/2 Alta study of
giroctocogene fitelparvovec for the treatment of severe hemophilia A. The data,
a summary of which is located below, showed that, as of the May 19, 2021 cutoff
date, for the four patients in the highest dose 3e13vg/kg cohort who had reached
104 weeks of follow-up, the mean Factor VIII, or FVIII, activity was 30.9% at
week 104 as measured by chromogenic assay. In this cohort, annualized bleeding
rate was zero for the first year after treatment and 0.9 throughout total
duration of follow-up. Giroctocogene fitelparvovec was generally well tolerated
as of the cutoff date. We and Pfizer will be presenting additional clinical data
from this study at ASH on December 12, 2021.
•We and Pfizer also announced that some of the patients treated in the Phase 3
AFFINE trial of giroctocogene fitelparvovec have experienced FVIII activity
greater than 150% following treatment. To date, none of these patients have
experienced thrombotic events and some have been treated with direct oral
anticoagulants to reduce thrombotic risk. Pfizer recently decided to voluntarily
pause screening and dosing of additional patients in this trial to implement a
proposed protocol amendment intended to provide guidelines for clinical
management of elevated FVIII levels. Subsequent to the voluntary pause, we also
recently learned that the FDA has put this trial on clinical hold. A clinical
hold is an order issued by the FDA to the trial sponsor to suspend an ongoing
clinical trial. We may not resume the AFFINE trial without FDA authorization. We
and Pfizer plan to share the proposed protocol amendment with the FDA and other
relevant review bodies and to respond to the clinical hold, after which we
expect to provide an update on the trial. We anticipate pivotal data readouts
for this trial to be based on full analyses of at least fifty patients. Over 50%
of the patients have been enrolled in the Phase 3 AFFINE trial.
•We have enrolled the first patient in our Phase 1/2 STEADFAST clinical study of
TX200 for the treatment of HLA-A2 mismatched kidney transplant rejection. We
expect the first two patients in this study to be dosed in the middle of 2022
following kidney transplantation. We continue to open study sites and screen
patients for this study.
•Biogen announced that the previously undisclosed neuromuscular pre-clinical
target in our collaboration is type 1 myotonic dystrophy (DM1).
•We recently completed and brought online our in-house cell therapy
manufacturing facility in our Brisbane, California headquarters and remain on
track to complete our in-house cell therapy manufacturing facility in Valbonne,
France by year-end.
•We appointed D. Mark McClung as Chief Operating Officer, who previously served
as Chief Business Officer.
Summary of Preliminary Results from the Phase 1/2 STAAR study of Isaralgagene
Civaparvovec
•STAAR is an ongoing Phase 1/2 multicenter, open-label, dose-ranging clinical
study to assess the safety and tolerability of a single infusion of isaralgagene
civaparvovec in Fabry disease patients ? 18 years of age.
                                       32
--------------------------------------------------------------------------------
  Table of     Contents
•Patients are infused intravenously with a single dose and followed for 52
weeks. A separate long-term follow-up study will follow this study. At least two
subjects will be dosed in each dose cohort, with a potential expansion in each
cohort.
•Patients who are on stable enzyme replacement therapy, or ERT, may withdraw ERT
after treatment in a controlled and monitored fashion at the discretion of the
patient and the investigator.
•The dose escalation phase includes males with classic Fabry disease. The study
will later be expanded to include females, as well as patients with
Fabry-associated cardiac and renal disease.
•The study's primary endpoint is incidence of treatment-emergent adverse events.
Additional safety evaluations include routine hematology, chemistry and liver
tests; vital signs; electrocardiogram; echocardiogram; serial alpha-fetoprotein
testing and magnetic resonance imaging, or MRI, of liver to monitor for
potential formation of any liver mass. Secondary endpoints include change from
baseline at specific time points over the one-year study period in
alpha-galactosidase A, or ?-Gal A, activity, globotriaosylceramide, or Gb3, and
lyso-Gb3 levels in plasma; frequency of ERT infusion; changes in renal function,
cardiac function and left ventricular mass, measured by cardiac MRI and rAAV2/6
vector clearance. Key exploratory endpoints include quality of life, Fabry
symptoms and neuropathic pain scores; and immune response to AAV6 capsid and
?-Gal A.
•As of the September 17, 2021 cutoff date, four patients, ranging in age from 22
to 48 years, were treated with isaralgagene civaparvovec. Two patients were
treated in Cohort 1 at the dose of 0.5e13 vg/kg and two patients were dosed in
Cohort 2 at the dose of 1e13 vg/kg.
•As of the September 17, 2021 cutoff date, isaralgagene civaparvovec was
generally well tolerated. One patient each in Cohorts 1 and 2 exhibited
treatment-related adverse events for a total of five events (hemoglobin
decreased, platelet count increased, rash and pyrexia), which were all
classified as mild (Grade 1). No treatment-related serious adverse events were
reported. No liver enzyme elevations requiring steroid treatment were recorded.
•Results of plasma a-Gal A activity for the four patients as of the cutoff date
are shown in the table below. All four patients exhibited above normal levels of
?-Gal A activity by Week 12 following treatment through 14 weeks for the most
recently treated patient and 52 weeks for the first patient treated. ?-Gal A
activity ranged from a 2-fold to 15-fold increase above mean normal activity
levels as of the last date of measurement. In the one patient with elevated
levels pre-treatment, plasma lyso-gb3 levels decreased by approximately 40% from
baseline within ten weeks after dosing through Week 32. The other three
patients, with low baseline levels of lyso-Gb3, maintained steady lyso-Gb3
levels through the cutoff date.
•Prophylactic steroids were not administered per the study protocol, and as of
the cutoff date, no patients had exhibited liver enzyme elevations requiring
steroid treatment.
•Several of the patients reported subjective improvements in quality-of-life
measures as of the cutoff date. Three of the four patients exhibited
improvements in anhidrosis (inability to sweat) or hypohydrosis (reduced ability
to sweat), a primary and common Fabry disease symptom.
•One of the four patients was on ERT, and one was formerly on ERT but had not
received ERT in the prior six months. Following treatment of these patients with
isaralgagene civaparvovec, investigators withdrew one of these patients from ERT
after the cutoff date and are planning to withdraw the other patient from ERT
based on the stability of ?-Gal A activity.
                                       33
--------------------------------------------------------------------------------
  Table of     Contents
Phase 1/2 STAAR Study: Plasma ?-Gal A activity

[[Image Removed: sgmo-20210930_g1.jpg]]
Biomarker results were evaluated from the 4 patients in the first 2 dose cohorts
(0.5e13 vg/kg and 1.0e13 vg/kg) as of the cutoff date of September 17, 2021.
(*) Fold change was calculated at last measured time point. ?-Gal A activity was
measured using a 3-hour reaction time and presented in nmol/h/mL. For Patients 1
and 4 this was sampled at ERT trough. Normal range and mean were determined
based on healthy male individuals.
Summary of Preliminary Safety and Efficacy Results from the Phase 1/2 PRECIZN-1
Study of SAR415536
•PRECIZN-1 is an ongoing first-in-human, open label, single arm, multi-site
study evaluating safety and tolerability of SAR445136 (n=8; aged 18-40 years),
with severe SCD across six U.S. sites.
•Eligible subjects underwent mobilization and apheresis with plerixafor.
Autologous hematopoietic stem and progenitor cells, or HSPCs, were transfected
ex vivo with ZFN messenger ribonucleic acid to manufacture SAR445136. A single
IV infusion was administered at least 72 hours after pre-conditioning with
busulfan.
•Subjects were monitored for stem cell engraftment and hematopoietic recovery,
adverse events, clinical and laboratory hemolysis markers, total hemoglobin and
fetal hemoglobin, percentage of F cells and sickle-cell related events
post-SAR445136 infusion.
•One subject failed to mobilize adequate cells. Of the seven subjects that
underwent mobilization and apheresis through the June 25, 2021 cutoff date, five
achieved successful target yields of HSPCs. One subject discontinued due to
intercurrent cholangitis. Baseline patient characteristics of the four patients
infused as of the cutoff date are in Table 1 below.
•All four patients improved clinically since SAR445136 infusion through the
cutoff date. Total hemoglobin stabilized at 9-10 g/dL by week 26 post SAR445136
infusion along with improvements in the clinical markers of hemolysis in all
four subjects. Percent fetal hemoglobin levels were 1-11% at screening,
increasing to 15-29% by week 13 in all four subjects, to 14-39% by week 26 in
the three subjects with at least 26 weeks of follow up, and persisting at 35% in
one subject with 65 weeks of follow up (see Figure 1 below). Percent F cells
increased to 49-94% in three subjects with at least 26 weeks of follow up,
persisting at 90% in one subject with 65 weeks of follow up. The fourth subject
had 87.5% F cells at 13 weeks of follow up.
•As of the June 25, 2021 cutoff date, SAR445136 was generally well tolerated
with no infusion related reactions. The adverse events reported were consistent
with plerixafor mobilization and busulfan myeloablation therapy. No adverse
events or serious adverse events were reported as related to SAR445136.
                                       34
--------------------------------------------------------------------------------
  Table of     Contents
Table 1. Baseline Characteristics and Clinical History
                                    Subject 103-002         Subject 100-001         Subject 102-001         Subject 103-003
Genotype                                 HbSB0                   HbSS                    HbSS                    HbSS
Gender                                  Female                  Female                   Male                    Male
Age at consent, years                     35                      20                      18                      26
Pain crises/2 years, n                    10                      22                       0                       6
Active chest syndrome events/2             2                       0                       4                       0
years, n
Regular, chronic RBC                     None                     Yes                     Yes                     Yes
transfusion therapy
Status as of the cutoff date       Week 65 completed       Week 26 completed       Week 26 completed       Week 13 completed

                                 No blood transfusions   No blood transfusions   No blood transfusions   No blood transfusions
                                   post engraftment        post engraftment

post engraftment post engraftment




[[Image Removed: sgmo-20210930_g2.jpg]]
HbF = Fetal hemoglobin, HbS = Sickle hemoglobin, HbA = Adult hemoglobin, HbA2 =
Hemoglobin A2
                                       35
--------------------------------------------------------------------------------
  Table of     Contents
Summary of Updated 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 19, 2021 cutoff date, one
patient in the highest dose cohort had not completed two years (104 weeks) of
follow up, resulting in patients having been followed for 95 to 195 weeks
overall.
•As of the May 19, 2021 cutoff date, the most commonly reported
treatment-related adverse events included elevated liver enzymes and
infusion-related reactions: increased alanine aminotransferase, or ALT (5/11
(45.5%) overall; 3/5 (60.0%) in the highest dose cohort), increased aspartate
aminotransferase, or AST (3/11 (27.3%) overall; 2/5 (40.0%) in the highest dose
cohort), pyrexia (3/11 (27.3%) overall; 3/5 (60.0%) in the highest dose cohort),
and tachycardia (2/11 (18.2%) overall; 2/5 (40.0%) in the highest dose cohort).
•Treatment-related serious adverse events 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 19, 2021 cutoff date; elevations in ALT were managed with a tapering course
of corticosteroids (median 58 days; range: 11-134 days), with maintenance of
clinically meaningful levels of 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 19, 2021 cutoff date, no patient in the study developed an
inhibitor to FVIII, and there have been no thrombotic events and no hepatic
masses detected.
•Patients in the highest dose cohort demonstrated FVIII activity as shown in the
table below through week 104 for the four patients in this cohort with available
data at week 104. In this cohort, the annualized bleeding rate, meaning the
number of all bleeding episodes starting three weeks after study drug 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 through week
104. In the highest dose cohort, two patients experienced a total of three
bleeding events (two traumatic; one unknown) necessitating treatment with
exogenous FVIII; one of these events occurred in a target joint. As of the
May 19, 2021 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, % Normal,                                                Study Week
Mean (SD)
Parameter                                Week 12             Week 24             Week 52             Week 78            Week 104
1-stage clotting                          110.9               107.5               97.9                79.4                46.4
                                         (36.4)              (79.2)              (135.5)             (73.3)              (37.0)
Chromogenic                               71.7                68.9                62.2                56.9                30.9
                                         (24.6)              (48.2)              (84.2)              (55.1)              (28.1)
Patients, n                                 5                   5                  4a                  4a                  4b

(a) There was one patient each that was unable to attend visits at Weeks 52 and Week 78. (b) One patient had not yet reached Week 104 of follow-up at the time of the data cut.




Estimated Impacts of Evolving COVID-19 Pandemic
We have experienced and continue to experience impacts from the 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, United Kingdom and locations of our clinical studies
and trials. We continue to conduct business operations pursuant to a modified
operating plan that includes enhanced workplace safety protocols and modified
working schedules. These protocols and modifications have slowed our
productivity and disrupted our business to a moderate degree and are likely to
continue doing so through the remainder of 2021 and possibly in 2022. For
example, we have experienced periodic short-term disruptions to our onsite
operations while addressing positive cases of COVID-19 by onsite workers, 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. Moreover,
from time to time, we have been required to reorganize and prioritize our
resources to mitigate moderate COVID-19
                                       36
--------------------------------------------------------------------------------
  Table of     Contents
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,
although we do not believe that the short-term disruptions to date have resulted
in any such impacts.
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 estimate that the opening of the first
clinical trial site in the United Kingdom for this study experienced a delay of
approximately one year due to the significant prevalence of COVID-19 in the
United Kingdom. Additionally, 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 prior to enrollment or dosing in the study.
Moreover, we have 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 estimate that these
challenges have set back our STAAR study timelines 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. We estimate that
these challenges set back our clinical study timeline by approximately three
months. While we have now enrolled the first patient in this study and expect to
dose the first two patients in this study in the middle of 2022, this timeline
could be revised 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 the extension of government orders or new
government orders 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
2021 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 and clinical programs. As of September 30, 2021, we had $519.0 million
in cash, cash equivalents, and marketable securities. Although we believe we are
well-capitalized currently, the effects of the evolving pandemic could result in
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, United Kingdom and other countries,
business closures or business disruptions and the effectiveness and timeliness
of actions taken in the United States, France, 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 has
resulted and may in the future result in the return of prior orders and
restrictions or new quarantine and shelter-in-place orders or other
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.
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
                                       37
--------------------------------------------------------------------------------
  Table of     Contents
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, Novartis and Sanofi,
certain expenses related to research and development activities will be
reimbursed to us. The reimbursement funds to be received from Biogen, Kite,
Novartis and Sanofi will be recognized as revenue as the related costs are
incurred and collection is reasonably assured.
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 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, 2021, 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 our 2020 Annual Report.
Results of Operations for the Three and Nine Months Ended September 30, 2021 and
2020
Revenues
                                              Three Months Ended September 30,                                                   Nine Months Ended September 30,
                                          (in thousands, except percentage values)                                           (in thousands, except percentage values)
                              2021                     2020              Change               %                   2021                     2020             Change               %
Revenues                $       28,563              $ 57,763          $ (29,200)            (51%)          $        82,715              $ 92,392          $ (9,677)            (10%)


Total revenues consisted of revenues from collaboration agreements and research
grants. We anticipate revenues over the next several years will be derived
primarily from our collaboration agreements with Biogen, Kite, Novartis, Pfizer
and Sanofi as we continue to recognize upfront and milestone payments received
under such agreements over time.
The decrease of $29.2 million in revenues for the three months ended
September 30, 2021, compared to the same period in 2020, was primarily
attributed to a decrease of $39.3 million of milestone fees and recognition of
upfront license fees related to our giroctocogene fitelparvovec and C9ORF72
collaboration agreements with Pfizer driven by completion of activities under
these collaborations in the fourth quarter of 2020, and a decrease of
$2.3 million in revenue related to our collaboration agreement with Sanofi,
primarily due to a change in estimate regarding project costs, resulting in a
decrease of proportional cumulative performance and a corresponding adjustment
to revenue under the agreement. These decreases were partially offset by
increases of $11.5 million related to our collaboration agreement with Novartis,
which became effective in July 2020, and an increase of $1.3 million in revenue
related to our collaboration agreement with Biogen.
The decrease of $9.7 million in revenues for the nine months ended September 30,
2021, compared to the same period in 2020, was primarily attributed to a
decrease of $46.7 million of milestone fees and recognition of upfront license
fees related to our giroctocogene fitelparvovec and C9ORF72 collaboration
agreements with Pfizer driven by completion of activities under these
collaborations in the fourth quarter of 2020, a decrease of $2.9 million in
research revenue related to our collaboration agreement with Kite, a decrease of
$2.5 million in revenue related to our collaboration agreement with Sanofi,
primarily due to a change in estimate regarding project costs, resulting in a
decrease of proportional cumulative performance and a corresponding adjustment
to revenue under the agreement, and a decrease of $1.3 million in revenue
related to sublicense fees under our agreement with Dow AgroSciences LLC. These
decreases were partially offset by increases of $29.0 million and $14.4 million
                                       38
--------------------------------------------------------------------------------
  Table of     Contents
due to the recognition of upfront license fees and research revenue under our
collaboration agreements with Novartis and Biogen, respectively.
Operating expenses
                                                       Three Months Ended September 30,                                                    Nine Months Ended September 30,
                                                   (in thousands, except percentage values)                                            (in

thousands, except percentage values)


                                          2021                 2020             Change               %                     2021                          2020             Change              %
Operating expenses:
Research and development           $        62,498          $ 45,287          $ 17,211              38%           $       179,018                    $ 128,289          $ 50,729             40%
General and administrative                  14,501            16,177            (1,676)            (10%)                   47,135                       50,223            (3,088)            (6%)
Total operating expenses           $        76,999          $ 61,464          $ 15,535              25%           $       226,153                    $ 178,512          $ 47,641             27%


Research and Development Expenses
Research and development expenses consisted primarily of compensation related
expenses, including stock-based compensation, laboratory supplies, preclinical
and clinical studies, manufacturing clinical supply, contracted research, and
allocated facilities and information technology expenses.
The increase of $17.2 million in research and development expenses for the three
months ended September 30, 2021, compared to the same period in 2020, was
primarily driven by a $7.6 million increase in preclinical, clinical and lab
supply expenses due to the timing of our trials and increased activity
attributed to our Biogen and Novartis collaborations, a $4.6 million increase in
manufacturing and overhead costs as we ramp up our internal manufacturing
operations, and a $3.3 million increase in compensation expense as a result of
increased headcount to support our programs, clinical trials and manufacturing
operations. Stock-based compensation expense included in research and
development expenses was $4.9 million and $3.6 million for the three months
ended September 30, 2021 and 2020, respectively.
The increase of $50.7 million in research and development expenses for the nine
months ended September 30, 2021, compared to the same period in 2020, was
primarily driven by a $21.3 million increase in preclinical, clinical and lab
supply expenses due to the timing of our trials and increased activity
attributed to our new collaborations, a $16.2 million increase in compensation
expense as a result of increased headcount to support our programs, clinical
trials and manufacturing operations, and a $12.6 million increase in
manufacturing and overhead costs as we ramp up our internal manufacturing
operations. Stock-based compensation expense included in research and
development expenses was $14.6 million and $10.0 million for the nine months
ended September 30, 2021 and 2020, 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. The full extent of the impact of the COVID-19 pandemic on our
business, operations and financial results will depend on numerous evolving
factors that we may not be able to accurately predict. 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 2020 Annual Report, as
supplemented by the risks and uncertainties described under "Risk Factors" in
Part II, Item 1A of this Quarterly Report.
                                       39
--------------------------------------------------------------------------------
  Table of     Contents
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, allocated facilities and
information technology expenses, and other general corporate expenses.
The decrease of $1.7 million in general and administrative expenses for the
three months ended September 30, 2021, compared to the same period in 2020, was
primarily due to a $0.9 million decrease in legal and professional fees, and a
decrease of $0.8 million due to reduced headcount and related compensation
costs. Stock-based compensation expense included in general and administrative
expenses was $2.9 million and $3.1 million for the three months ended
September 30, 2021 and 2020, respectively.
The decrease of $3.1 million in general and administrative expenses for the nine
months ended September 30, 2021, compared to the same period in 2020, was
primarily due to a $2.6 million decrease in legal and professional fees, and a
$0.3 million decrease in allocated facility overhead costs. Stock-based
compensation expense included in general and administrative expenses was
$10.2 million and $9.1 million for the nine months ended September 30, 2021 and
2020, 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, decreased by $1.6 million for the three months
ended September 30, 2021, compared to the same period in 2020, primarily due to
a decrease of $1.4 million as a result of fluctuations in foreign exchange
rates, and a decrease of $0.7 million in interest income due to lower portfolio
yields as a result of decrease in interest rates, partially offset by an
increase of $0.5 million in research tax credits earned by Sangamo France.
Interest and other income, net, decreased by $2.9 million for the nine months
ended September 30, 2021, compared to the same period in 2020, primarily due to
a decrease of $3.2 million in interest income due to lower portfolio yields as a
result of decrease in interest rates, and a decrease of $1.7 million as a result
of fluctuations in foreign exchange rates, partially offset by an increase of
$2.0 million in research tax credits earned by Sangamo France.
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, 2021, we had cash, cash equivalents, and marketable
securities totaling $519.0 million compared to $692.0 million as of December 31,
2020. Our most significant use of capital was for employee compensation and
external research and development expenses, such as 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,
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, or Jefferies, 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, 2021, we sold 2,007,932 shares of our common stock under the
sales agreement for net proceeds of approximately $27.1 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, strategic partnerships and research grants, will
be adequate to fund our currently planned operations through at least the next
12 months from the date the Condensed Consolidated Financial Statements are
issued. During this period of uncertainty and volatility related to the COVID-19
pandemic, we will continue to monitor our liquidity.
Cash Flows
Operating activities
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
                                       40
--------------------------------------------------------------------------------
  Table of     Contents
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.
Net cash provided by operating activities was $174.2 million for the nine months
ended September 30, 2020, primarily reflecting an increase in deferred revenues
of $235.6 million due to cash received in connection with the Biogen
collaboration agreement and the Novartis collaboration agreement, and $23.1
million of non-cash expenses related to stock-based compensation and
depreciation, partially offset by our net loss of $80.4 million.
Investing activities
Net cash provided by investing activities was $195.6 million for the nine months
ended September 30, 2021, mostly related to net maturities, sales and purchases
of marketable securities, partially offset by $20.4 million purchases of
property and equipment. Net cash used in investing activities for the nine
months ended September 30, 2020 was $141.2 million, mostly related to net
maturities and purchases of marketable securities, and purchases of property and
equipment.
Financing activities
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. Net cash
provided by financing activities for the nine months ended September 30, 2020
was $146.2 million, primarily reflecting the $145.4 million estimated fair value
of the shares issued to Biogen offset by $2.9 million of issuance costs related
to the issuance, and an increase of $4.2 million related to proceeds from the
exercise of stock options and restricted stock units and purchases under the
employee stock purchase plan.
Operating Capital and Capital Expenditure Requirements
We anticipate continuing to incur operating losses for at least the next several
years. 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, strategic partners and research grants,
will be adequate to fund our currently planned operations through at least the
next 12 months from the date the Condensed Consolidated Financial Statements are
issued. Although we believe we are well capitalized currently, the effects of
the ongoing COVID-19 pandemic 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 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 decide, from time to time, to raise 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 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;
                                       41

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


  Table of     Contents
•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.
Off-Balance Sheet Arrangements
We do not have any off-balance sheet arrangements as defined in
Item 303(a)(4)(ii) of Regulation S-K.
Contractual Obligations
Our future minimum contractual obligations as of December 31, 2020 were reported
in the 2020 Annual Report. Other than as described below, during the nine months
ended September 30, 2021, there have been no other material changes outside the
ordinary course of our business from the contractual obligations previously
disclosed in our 2020 Annual Report.
In January 2021, we also entered into a new lease to acquire approximately
5,800 square feet of research and office space in Valbonne, France that expires
in January 2030. The contractual obligations during the lease term are
approximately $0.8 million.
In October 2021, we entered into an agreement to extend the lease of our
research and office space in Richmond, California. For further information on
the lease, see Note 11 in the accompanying Notes to Condensed Consolidated
Financial Statements.

© Edgar Online, source Glimpses