UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

WASHINGTON, DC 20549

FORM 10-Q

(Mark One)

QUARTERLY REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

QUARTERLY REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

For the quarterly period ended March 31, 20212022

OR

TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

For the transition period from to

Commission File Number: 001-37785

Reata Pharmaceuticals, Inc.

(Exact Name of Registrant as Specified in its Charter)

 

Delaware

11-3651945

(State or other jurisdiction of

incorporation or organization)

(I.R.S. Employer
Identification No.)

5320 Legacy Drive
Plano, Texas

75024

(Address of principal executive offices)

(Zip Code)

(972) (972) 865-2219

(Registrant’s telephone number, including area code)

Securities registered pursuant to Section 12(b) of the Securities Exchange Act of 1934:

Title of each class

Trading

Symbol(s)

Name of each exchange on which registered

Class A Common Stock, Par Value $0.001 Per Share

RETA

NASDAQ Global Market

Indicate by check mark whether the registrant (1) has filed all reports required to be filed by Section 13 or 15(d) of the Securities Exchange Act of 1934 during the preceding 12 months (or for such shorter period that the registrant was required to file such reports), and (2) has been subject to such filing requirements for the past 90 days. Yes No

Indicate by check mark whether the registrant has submitted electronically every Interactive Data File required to be submitted pursuant to Rule 405 of Regulation S-T (§ 232.405 of this chapter) during the preceding 12 months (or for such shorter period that the registrant was required to submit such files). Yes No

Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, an emerging growth company, or a smaller reporting company. See the definitions of “large accelerated filer,” “accelerated filer,” “smaller reporting company,” and “emerging growth company” in Rule 12b-2 of the Exchange Act.

Large accelerated filer

 

 

Accelerated filer

 

Non-accelerated filer

 

 

Smaller reporting company

 

Emerging growth company

If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act.

Indicate by check mark whether the registrant is a shell company (as defined in Rule 12b-2 of the Exchange Act). Yes No

As of April 30, 2021,May 4, 2022, the registrant had 31,368,32031,543,735 shares of Class A common stock, $0.001 par value per share, and 4,909,4924,919,249 shares of Class B common stock, $0.001 par value per share, outstanding.


TABLE OF CONTENTS

-

 

Page

CAUTIONARY NOTE REGARDING FORWARD-LOOKING STATEMENTS

1

DEFINED TERMS

3

PART I.

FINANCIAL INFORMATION

Item 1.

Financial Statements (Unaudited)

4

 

Consolidated Balance Sheets

4

 

Consolidated Statements of Operations

5

 

Consolidated Statements of Stockholders’ Equity (Deficit)

6

 

Consolidated Statements of Cash Flows

7

 

Notes to Unaudited Consolidated Financial Statements

8

Item 2.

Management’s Discussion and Analysis of Financial Condition and Results of Operations

16

Item 3.

Quantitative and Qualitative Disclosures About Market Risk

3435

Item 4.

Controls and Procedures

35

PART II.

OTHER INFORMATION

Item 1.

Legal Proceedings

3536

Item 1A.

Risk Factors

3536

Item 2.

Unregistered Sales of Equity Securities and Use of Proceeds

3536

Item 3.

Defaults Upon Senior Securities

36

Item 4.

Mine Safety Disclosures

36

Item 5.

Other Information

36

Item 6.

Exhibits

37

Signatures

38

i


CAUTIONARY NOTE REGARDING FORWARD-LOOKING STATEMENTS

This Quarterly Report on Form 10-Q contains forward-looking statements that involve substantial risks and uncertainties. We make such forward-looking statements pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995 and other federal securities laws. In this Quarterly Report on Form 10-Q, all statements, other than statements of historical or present facts, including statements regarding our future financial condition, future revenues, projected costs, prospects, business strategy, and plans and objectives of management for future operations, are forward-looking statements. In some cases, you can identify forward-looking statements by terminology such as “believe,” “will,” “may,” “might,” “estimate,” “continue,” “anticipate,” “intend,” “target,” “project,” “model,” “should,” “would,” “plan,” “expect,” “predict,” “could,” “seek,” “goals,” “potential,” and similar terms or expressions that concern our expectations, strategy, plans, or intentions. These forward-looking statements include, but are not limited to, statements about:

our expectations regarding the timing, costs, conduct, and outcome of our clinical trials, including statements regarding the timing of the initiation and availability of data from such trials;
the timing and likelihood of regulatory filings and approvals for our product candidates;
whether regulatory authorities determine that additional trials or data are necessary in order to accept a new drug application for review and/or approval;
our ability to obtain funding for our operations, including funding necessary to complete further development and commercialization of our product candidates;
our plans to research, develop, and commercialize our product candidates;
the manufacturing, supply, and commercialization of our product candidates, if approved;
the rate and degree of market acceptance of our product candidates;
our expectations regarding the potential market size and the size of the patient populations for our product candidates, if approved for commercial use, and the potential market opportunities for commercializing our product candidates;
the success of competing therapies that are or may become available;
our expectations regarding our ability to obtain and maintain intellectual property protection for our product candidates;
the ability to license additional intellectual property relating to our product candidates and to comply with our existing license agreements;
our ability to maintain and establish relationships with third parties, such as contract research organizations (CROs), contract manufacturing organizations, suppliers, and distributors;
our ability to maintain and establish collaborators with development, regulatory, and commercialization expertise;
our ability to attract and retain key scientific or management personnel;
our ability to grow our organization and increase the size of our facilities to meet our anticipated growth;
the accuracy of our estimates regarding expenses, future revenue, capital requirements, and needs for additional financing;
our expectations related to the use of our available cash;
our ability to develop, acquire, and advance product candidates into, and successfully complete, clinical trials;

1


the initiation, timing, progress, and results of future preclinical studies and clinical trials, and our research and development programs;
the impact of governmental laws and regulations and regulatory developments in the United States and foreign countries;
developments and projections relating to our competitors and our industry; and
the impact of the coronavirus disease (COVID-19) on our clinical trials, our supply chain, and our operations; and
other risks and uncertainties, including those described under the heading “Risk Factors” included in our most recent Annual Report on Form 10-K for the year ended December 31, 2021, filed with the U.S. Securities and Exchange Commission (SEC) on February 28, 2022.

our expectations regarding the timing, costs, conduct, and outcome of our clinical trials, including statements regarding the timing of the initiation and availability of data from such trials;

the timing and likelihood of regulatory filings and approvals for our product candidates;

whether regulatory authorities determine that additional trials or data are necessary in order to accept a new drug application for review and/or approval;

our ability to obtain funding for our operations, including funding necessary to complete further development and commercialization of our product candidates;

our plans to research, develop, and commercialize our product candidates;

the commercialization of our product candidates, if approved;

the rate and degree of market acceptance of our product candidates;

our expectations regarding the potential market size and the size of the patient populations for our product candidates, if approved for commercial use, and the potential market opportunities for commercializing our product candidates;

the success of competing therapies that are or may become available;

our expectations regarding our ability to obtain and maintain intellectual property protection for our product candidates;

the ability to license additional intellectual property relating to our product candidates and to comply with our existing license agreements;

our ability to maintain and establish relationships with third parties, such as contract research organizations (CROs), contract manufacturing organizations, suppliers, and distributors;

our ability to maintain and establish collaborators with development, regulatory, and commercialization expertise;

our ability to attract and retain key scientific or management personnel;

our ability to grow our organization and increase the size of our facilities to meet our anticipated growth;

the accuracy of our estimates regarding expenses, future revenue, capital requirements, and needs for additional financing;

our expectations related to the use of our available cash;

our ability to develop, acquire, and advance product candidates into, and successfully complete, clinical trials;


the initiation, timing, progress, and results of future preclinical studies and clinical trials, and our research and development programs;

the impact of governmental laws and regulations and regulatory developments in the United States and foreign countries;

developments and projections relating to our competitors and our industry;

the impact of the coronavirus disease (COVID-19) on our clinical trials, our supply chain, and our operations; and

other risks and uncertainties, including those described under the heading “Risk Factors” included in our most recent Annual Report on Form 10-K for the year ended December 31, 2020, filed with the U.S. Securities and Exchange Commission (SEC) on March 1, 2021.

Any forward-looking statements in this Quarterly Report on Form 10-Q reflect our current views with respect to future events or to our future financial performance 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, performance, or achievements expressed or implied by these forward-looking statements. Given these uncertainties, you should not place undue reliance on these forward-looking statements.

You should read this Quarterly Report on Form 10-Q and the documents that we have filed as exhibits to this Quarterly Report on Form 10-Q completely and with the understanding that our actual future results may be materially different from what we expect. Except as required by law, we assume no obligation to update or revise these forward-looking statements for any reason, even if new information becomes available in the future.


2


DEFINED TERMS

Unless the context requires otherwise, references to “Reata,” “the Company,” “we,” “us,” or “our” in this Quarterly Report on Form 10-Q refer to Reata Pharmaceuticals, Inc. and its subsidiaries. We also have used several other terms in this Quarterly Report on Form 10-Q, most of which are explained or defined below.

Abbreviated Term

Defined Term

AbbVie

AbbVie Inc.

ADPKD

Autosomal dominant polycystic kidney disease

AEADL

Adverse eventActivities of Daily Living

ASCAE

Accounting Standards CodificationAdverse event

ASUALS

Accounting Standards UpdateAmyotrophic lateral sclerosis

ATP

Adenosine triphosphate production

Bardoxolonebardoxolone

Bardoxolone methyl

BXLS

Blackstone Life Sciences, LLC

CARES ActCKD

Coronavirus Aid, Relief, and Economic Security ActChronic kidney disease

CKDCMC

Chronic kidney diseaseChemistry manufacturing controls

COVID-19

Coronavirus disease

CROCRL

Contract research organizationComplete Response Letter

DPNPCRO

Contract research organization

DPNP

Diabetic peripheral neuropathic pain

eGFR

Estimated glomerular filtration rate

EMA

European Medicines Agency

ESKD

End stage kidney disease

Exchange Act

Securities Exchange Act of 1934

FA

Friedreich’s ataxia

FASBFDA

Financial Accounting Standards Board

FDA

United States Food and Drug Administration

FSGSGFR

Focal segmental glomerulosclerosisGlomerular filtration rate

GFRKyowa Kirin

Glomerular filtration rate

IgAN

IgA nephropathy

ITT

Intent to treat

KKC

Kyowa Kirin Co., Ltd.

MAALTIP Plan

Marketing Authorization ApplicationSecond Amended and Restated Long Term Incentive Plan

mFARSMAA

Marketing Authorization Application

mFARS

Modified Friedreich’s Ataxia Rating Scale

mITTNDA

Modified ITTNew Drug Application

NDAPGIC

New Drug ApplicationPatient global impression of change

PAHPK

Pulmonary arterial hypertensionPharmacokinetic

PDUFARegistrational trial

Prescription Drug User Fee Act

PK

Pharmacokinetic

Registrational trial

An adequate and well-controlled trial designed to be sufficient to apply for regulatory

approval of a drug candidate, although notwithstanding the Company’s design a

regulatory agency may determine that further clinical studies or data are required

RSU

Restricted Stock Unit

SAE

Serious adverse event

SEC

U.S. Securities and Exchange Commission

T1D CKDU.S. GAAP

Type 1 diabetic CKD

T2D CKD

Type 2 diabetic CKD

UACR

Urinary albumin-to-creatinine ratioAccounting principles generally accepted in the United States


3


PART I - FINANCIAL INFORMATION

Item 1. Financial Statements.

Reata Pharmaceuticals, Inc.

Consolidated Balance Sheets

(in thousands, except share data)

 

March 31, 2021

 

 

December 31, 2020

 

 

March 31, 2022

 

 

December 31, 2021

 

 

(unaudited)

 

 

 

 

 

 

(unaudited)

 

 

 

Assets

 

 

 

 

 

 

 

 

 

 

 

 

 

Cash and cash equivalents

 

$

777,624

 

 

$

818,150

 

 

$

531,979

 

$

590,258

 

Prepaid expenses and other current assets

 

 

5,646

 

 

 

6,960

 

 

 

5,357

 

 

 

6,217

 

Income tax receivable

 

 

22,250

 

 

 

22,228

 

Total current assets

 

 

805,520

 

 

 

847,338

 

 

537,336

 

596,475

 

Property and equipment, net

 

 

4,922

 

 

 

4,912

 

 

11,202

 

11,604

 

Operating lease right-of-use-assets

 

131,178

 

126,777

 

Other assets

 

 

4,642

 

 

 

5,348

 

 

 

152

 

 

 

160

 

Total assets

 

$

815,084

 

 

$

857,598

 

 

$

679,868

 

 

$

735,016

 

Liabilities and stockholders’ equity

 

 

 

 

 

 

 

 

 

 

 

 

 

Accounts payable

 

 

8,255

 

 

 

4,790

 

 

9,015

 

13,505

 

Accrued direct research liabilities

 

 

12,618

 

 

 

14,023

 

 

14,930

 

14,249

 

Other current liabilities

 

 

14,443

 

 

 

22,264

 

 

13,778

 

21,450

 

Payable to collaborators

 

 

75,150

 

 

 

73,437

 

Operating lease liabilities, current

 

5,142

 

3,142

 

Deferred revenue

 

 

3,893

 

 

 

4,688

 

 

 

755

 

 

 

1,648

 

Total current liabilities

 

 

114,359

 

 

 

119,202

 

 

43,620

 

53,994

 

Other long-term liabilities

 

 

5,013

 

 

 

5,511

 

 

5

 

0

 

Operating lease liabilities, noncurrent

 

136,445

 

132,891

 

Liability related to sale of future royalties, net

 

 

326,379

 

 

 

315,454

 

 

 

372,013

 

 

 

362,142

 

Total noncurrent liabilities

 

 

331,392

 

 

 

320,965

 

 

508,463

 

495,033

 

Commitments and contingencies

 

 

 

 

 

 

 

 

 

 

 

 

 

Stockholders’ equity:

 

 

 

 

 

 

 

 

 

 

 

 

 

Common stock A, $0.001 par value:

500,000,000 shares authorized; issued and outstanding – 31,360,256 and

31,109,154 at March 31, 2021 and December 31, 2020, respectively

 

 

31

 

 

 

31

 

Common stock B, $0.001 par value:

150,000,000 shares authorized; issued and outstanding – 4,909,554 and

5,044,931 at March 31, 2021 and December 31, 2020, respectively

 

 

5

 

 

 

5

 

Common stock A, $0.001 par value:
500,000,000 shares authorized; issued and outstanding – 31,525,514 and
31,478,197 at March 31, 2022 and December 31, 2021, respectively

 

31

 

31

 

Common stock B, $0.001 par value:
150,000,000 shares authorized; issued and outstanding – 4,919,249 and
4,919,249 at March 31, 2022 and December 31, 2021, respectively

 

5

 

5

 

Additional paid-in capital

 

 

1,394,997

 

 

 

1,375,640

 

 

1,457,222

 

1,441,584

 

Accumulated deficit

 

 

(1,025,700

)

 

 

(958,245

)

 

 

(1,329,473

)

 

 

(1,255,631

)

Total stockholders’ equity

 

 

369,333

 

 

 

417,431

 

 

 

127,785

 

 

 

185,989

 

Total liabilities and stockholders’ equity

 

$

815,084

 

 

$

857,598

 

 

$

679,868

 

 

$

735,016

 

 

See accompanying notes.


4


Reata Pharmaceuticals, Inc.

Unaudited Consolidated Statements of Operations

(in thousands, except share and per share data)

 

Three Months Ended

 

 

Three Months Ended

 

 

March 31

 

 

March 31

 

 

2021

 

 

2020

 

 

2022

 

 

2021

 

Collaboration revenue

 

 

 

 

 

 

 

 

 

 

 

 

 

License and milestone

 

$

795

 

 

$

1,169

 

 

$

893

 

$

795

 

Other revenue

 

 

149

 

 

 

184

 

 

 

21

 

 

 

149

 

Total collaboration revenue

 

 

944

 

 

 

1,353

 

 

914

 

944

 

Expenses

 

 

 

 

 

 

 

 

 

 

 

 

 

Research and development

 

 

34,880

 

 

 

47,653

 

 

39,804

 

34,880

 

General and administrative

 

 

20,704

 

 

 

20,787

 

 

24,841

 

20,704

 

Depreciation

 

 

274

 

 

 

278

 

 

 

308

 

 

 

274

 

Total expenses

 

 

55,858

 

 

 

68,718

 

 

64,953

 

55,858

 

Other income (expense), net

 

 

(12,556

)

 

 

(3,814

)

 

 

(9,772

)

 

 

(12,556

)

Loss before taxes on income

 

 

(67,470

)

 

 

(71,179

)

 

(73,811

)

 

(67,470

)

Benefit from (provision for) taxes on income

 

 

15

 

 

 

22,240

 

 

 

(31

)

 

 

15

 

Net loss

 

$

(67,455

)

 

$

(48,939

)

 

$

(73,842

)

 

$

(67,455

)

Net loss per share—basic and diluted

 

$

(1.86

)

 

$

(1.47

)

 

$

(2.03

)

 

$

(1.86

)

Weighted-average number of common shares used in

net loss per share basic and diluted

 

 

36,203,631

 

 

 

33,222,085

 

 

36,412,621

 

36,203,631

 

See accompanying notes.


5


Reata Pharmaceuticals, Inc.

Unaudited Consolidated Statements of Stockholders’ Equity

(in thousands, except share and per share data)

 

Three Months Ended March 31, 2021

 

 

Common Stock A

 

 

Common Stock B

 

 

Additional

Paid-In

 

 

Total

Accumulated

 

 

Total

Stockholders’

 

 

Three Months Ended March 31, 2022

 

 

Shares

 

 

Amount

 

 

Shares

 

 

Amount

 

 

Capital

 

 

Deficit

 

 

Equity

 

 

Common Stock A

 

 

Common Stock B

 

 

Additional
Paid-In

 

Total
Accumulated

 

Total
Stockholders’

 

Balance at December 31, 2020

 

 

31,109,154

 

 

$

31

 

 

 

5,044,931

 

 

$

5

 

 

$

1,375,640

 

 

$

(958,245

)

 

$

417,431

 

 

Shares

 

 

Amount

 

 

Shares

 

 

Amount

 

 

Capital

 

 

Deficit

 

 

Equity

 

Balance at December 31, 2021

 

 

31,478,197

 

 

$

31

 

 

 

4,919,249

 

 

$

5

 

 

$

1,441,584

 

 

$

(1,255,631

)

 

$

185,989

 

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(67,455

)

 

 

(67,455

)

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(73,842

)

 

 

(73,842

)

Compensation expense

related to stock options

 

 

 

 

 

 

 

 

 

 

 

 

 

 

14,679

 

 

 

 

 

 

14,679

 

 

 

 

 

 

 

 

 

 

 

 

 

 

15,444

 

 

 

 

 

 

15,444

 

Exercise of options

 

 

 

 

 

 

 

 

112,423

 

 

 

 

 

 

4,678

 

 

 

 

 

 

4,678

 

 

 

 

 

 

 

 

9,375

 

 

 

 

 

 

194

 

 

 

 

 

 

194

 

Issuance of common stock upon

vesting of restricted stock units

 

 

 

 

 

 

 

 

3,302

 

 

 

 

 

 

 

 

 

 

 

 

 

 

35,107

 

 

 

 

 

 

2,835

 

 

 

 

 

 

 

 

 

 

 

 

 

Conversion of common

stock Class B to Class A

 

 

251,102

 

 

 

 

 

 

(251,102

)

 

 

 

 

 

 

 

 

 

 

 

 

 

 

12,210

 

 

 

 

 

 

(12,210

)

 

 

 

 

 

 

 

 

 

 

 

 

Balance at March 31, 2021

 

 

31,360,256

 

 

$

31

 

 

 

4,909,554

 

 

$

5

 

 

$

1,394,997

 

 

$

(1,025,700

)

 

$

369,333

 

Balance at March 31, 2022

 

 

31,525,514

 

 

$

31

 

 

 

4,919,249

 

 

$

5

 

 

$

1,457,222

 

 

$

(1,329,473

)

 

$

127,785

 

 

 

Three Months Ended March 31, 2020

 

 

 

Common Stock A

 

 

Common Stock B

 

 

Additional

Paid-In

 

 

Total

Accumulated

 

 

Total

Stockholders’

 

 

 

Shares

 

 

Amount

 

 

Shares

 

 

Amount

 

 

Capital

 

 

Deficit

 

 

Equity

 

Balance at December 31, 2019

 

 

27,878,550

 

 

$

28

 

 

 

5,318,157

 

 

$

5

 

 

$

967,317

 

 

$

(710,493

)

 

$

256,857

 

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(48,939

)

 

 

(48,939

)

Compensation expense

   related to stock options

 

 

 

 

 

 

 

 

 

 

 

 

 

 

19,307

 

 

 

 

 

 

19,307

 

Exercise of options

 

 

 

 

 

 

 

 

40,216

 

 

 

 

 

 

1,422

 

 

 

 

 

 

1,422

 

Conversion of common

   stock Class B to Class A

 

 

288,102

 

 

 

 

 

 

(288,102

)

 

 

 

 

 

 

 

 

 

 

 

 

Balance at March 31, 2020

 

 

28,166,652

 

 

$

28

 

 

 

5,070,271

 

 

$

5

 

 

$

988,046

 

 

$

(759,432

)

 

$

228,647

 

 

 

Three Months Ended March 31, 2021

 

 

 

Common Stock A

 

 

Common Stock B

 

 

Additional
Paid-In

 

 

Total
Accumulated

 

 

Total
Stockholders’

 

 

 

Shares

 

 

Amount

 

 

Shares

 

 

Amount

 

 

Capital

 

 

Deficit

 

 

Equity

 

Balance at December 31, 2020

 

 

31,109,154

 

 

$

31

 

 

 

5,044,931

 

 

$

5

 

 

$

1,375,640

 

 

$

(958,245

)

 

$

417,431

 

Net loss

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

(67,455

)

 

 

(67,455

)

Compensation expense
   related to stock options

 

 

 

 

 

 

 

 

 

��

 

 

 

 

14,679

 

 

 

 

 

 

14,679

 

Exercise of options

 

 

 

 

 

 

 

 

112,423

 

 

 

 

 

 

4,678

 

 

 

 

 

 

4,678

 

Issuance of common stock upon
   vesting of restricted stock units

 

 

 

 

 

 

 

 

3,302

 

 

 

 

 

 

 

 

 

 

 

 

 

Conversion of common
   stock Class B to Class A

 

 

251,102

 

 

 

 

 

 

(251,102

)

 

 

 

 

 

 

 

 

 

 

 

 

Balance at March 31, 2021

 

 

31,360,256

 

 

$

31

 

 

 

4,909,554

 

 

$

5

 

 

$

1,394,997

 

 

$

(1,025,700

)

 

$

369,333

 

See accompanying notes.


6


Reata Pharmaceuticals, Inc.

Unaudited Consolidated Statements of Cash Flows

(in thousands)

 

Three Months Ended

 

 

Three Months Ended

 

 

March 31

 

 

March 31

 

 

2021

 

 

2020

 

 

2022

 

 

2021

 

Operating activities

 

 

 

 

 

 

 

 

 

 

 

 

 

Net loss

 

$

(67,455

)

 

$

(48,939

)

 

$

(73,842

)

 

$

(67,455

)

Adjustments to reconcile net loss to net cash used in operating activities:

 

 

 

 

 

 

 

 

 

 

 

 

 

Depreciation

 

 

274

 

 

 

278

 

 

308

 

274

 

Amortization of debt issuance costs and imputed interest

 

 

1,714

 

 

 

489

 

 

0

 

1,714

 

Non-cash interest expense on liability related to sale of future royalty

 

 

10,925

 

 

 

0

 

 

9,871

 

10,925

 

Stock-based compensation expense

 

 

14,679

 

 

 

19,307

 

 

15,444

 

14,679

 

Changes in operating assets and liabilities:

 

 

 

 

 

 

 

 

 

 

 

 

 

Income tax receivable and payable

 

 

(22

)

 

 

(22,218

)

 

0

 

(22

)

Prepaid expenses and other current assets and other assets

 

 

1,330

 

 

 

809

 

Prepaid expenses, other current assets and other assets

 

877

 

1,330

 

Accounts payable

 

 

3,629

 

 

 

10,668

 

 

(4,525

)

 

3,629

 

Accrued direct research, other current and long-term liabilities

 

 

(9,290

)

 

 

(1,976

)

 

(8,914

)

 

(9,301

)

Payable to collaborators

 

 

0

 

 

 

1,578

 

Operating lease obligations

 

3,489

 

11

 

Deferred revenue

 

 

(795

)

 

 

(1,169

)

 

 

(893

)

 

 

(795

)

Net cash used in operating activities

 

 

(45,011

)

 

 

(41,173

)

 

 

(58,185

)

 

 

(45,011

)

Investing activities

 

 

 

 

 

 

 

 

 

 

 

 

 

Purchases of property and equipment

 

 

(193

)

 

 

(85

)

 

 

(288

)

 

 

(193

)

Net cash used in investing activities

 

 

(193

)

 

 

(85

)

 

 

(288

)

 

 

(193

)

Financing activities

 

 

 

 

 

 

 

 

 

 

 

 

 

Exercise of options

 

 

4,678

 

 

 

1,422

 

 

 

194

 

 

 

4,678

 

Net cash provided by financing activities

 

 

4,678

 

 

 

1,422

 

 

 

194

 

 

 

4,678

 

Net decrease in cash and cash equivalents

 

 

(40,526

)

 

 

(39,836

)

 

(58,279

)

 

(40,526

)

Cash and cash equivalents at beginning of year

 

 

818,150

 

 

 

664,324

 

 

 

590,258

 

 

 

818,150

 

Cash and cash equivalents at end of period

 

$

777,624

 

 

$

624,488

 

 

$

531,979

 

 

$

777,624

 

Supplemental disclosures

 

 

 

 

 

 

 

 

Cash paid for interest

 

$

0

 

 

$

3,174

 

Non-cash activity:

 

 

 

 

 

 

 

 

 

 

 

 

 

Right-of-use assets obtained in exchange for lease obligations

 

$

4,885

 

$

0

 

Purchases of equipment in accounts payable, accrued direct research, other current, and long-term liabilities

 

$

2,523

 

 

$

1,398

 

 

$

2,258

 

$

28

 

Acquisition of property and equipment through tenant improvement allowance

 

$

0

 

$

2,495

 

See accompanying notes.


7


Reata Pharmaceuticals, Inc.

Notes to Unaudited Consolidated Financial Statements

1. Description of Business

The Company’sReata Pharmaceuticals, Inc.’s (Reata, the Company, we, us, or our) mission is to identify, develop, and commercialize innovative therapies that change patients’ lives for the better. The Company focuses on small-molecule therapeutics with novel mechanisms of action for the treatment of severe, life-threatening diseases with few or no approved therapies. The Company’s lead programs are omaveloxolone in a rare neurological disease called Friedreich’s ataxia (FA) and bardoxolone methyl (bardoxolone) in rare forms of chronic kidney disease (CKD) and a rare neurological disease.  The Company announced positive topline data from registrational trials for both. Both of itsthe Company’s lead product candidates bardoxolone methyl (bardoxolone) in patients with CKD caused by Alport syndrome and omaveloxolone in patients with a neurological disorder called Friedreich’s ataxia (FA).  Both bardoxolone and omaveloxolone activate the transcription factor Nrf2 to normalize mitochondrial function, restore redox balance, and resolve inflammation. Because mitochondrial dysfunction, oxidative stress, and inflammation are features of many diseases, the Company believes omaveloxolone, bardoxolone, omaveloxolone, and our next-generation Nrf2 activators have many potential clinical applications. Reata possesses exclusive, worldwide rights to develop, manufacture, and commercialize omaveloxolone, bardoxolone, omaveloxolone, and our next-generation Nrf2 activators, excluding certain Asian markets for bardoxolone in certain indications, which are licensed to Kyowa Kirin Co., Ltd. (KKC)(Kyowa Kirin). In addition, we are developing RTA 901, the lead product candidate from our Hsp90 modulator program, in neurological indications. We are the exclusive licensee of RTA 901 and have worldwide commercial rights.

The Company’s consolidated financial statements include the accounts of all majority-owned subsidiaries. Accordingly, the Company’s share of net earnings and losses from these subsidiaries is included in the consolidated statements of operations. Intercompany profits, transactions, and balances have been eliminated in consolidation.consolidation.

Prior period reclassifications

Certain prior period amounts in the consolidated financial statements have been reclassified to conform to the current period presentation. Specifically, Operating lease obligations have been reclassed out of Accrued direct research, other current and long-term liabilities in prior periods to conform with the current period presentation on the consolidated statements of cash flows.

2. Summary of Significant Accounting Policies

Basis of Presentation

The accompanying unaudited consolidated financial statements have been prepared in accordance with accounting principles generally accepted in the United States (U.S. GAAP) for interim financial information and with the instructions to Form 10-Q and Article 10 of Regulation S-X. Accordingly, they do not include all of the information and notes required by U.S. GAAP for complete financial statements. In the opinion of management, all adjustments (consisting of normal recurring adjustments) considered necessary for a fair presentation have been included. Operating results for the three months ended March 31, 20212022 are not necessarily indicative of the results that may be expected for the year ending December 31, 2021.2022. The consolidated balance sheet at December 31, 2020,2021, has been derived from the audited consolidated financial statements at that date but does not include all of the information and footnotes required by U.S. GAAP for complete financial statements. For further information, refer to the annual consolidated financial statements and footnotes thereto of the Company.

Summary of Significant Accounting Policies

The significant accounting policies used in the preparation of these condensed consolidated financial statements for the three months ended March 31, 20212022 are consistent with those discussed in Note 2 to the consolidated financial statements in the Company’s Annual Report on Form 10-K for the year ended December 31, 2020.2021.

Recently Adopted Accounting Pronouncements

In December 2019, the FASB issued ASU 2019-12, Income Taxes (Topic 740), Simplifying the Accounting for Income Taxes.  The FASB issued this update as part of its Simplification Initiative to improve areas of U.S. GAAP and reduce cost and complexity while maintaining usefulness.  The main provision that impacts the Company is the removal of the exception to the incremental approach of intra-period tax allocation when there is a loss from continuing operations and income or gain from other items.  ASU 2019-12 is effective for annual periods, and interim periods within those annual periods, beginning after December 15, 2020.  The Company adopted this


standard on January 1, 2021 and its adoption did not have material impact to the Company’s consolidated financial statements and related disclosure.

8


3. Collaboration Agreements

KKCSubsequent to the 2019 reacquisition of certain rights originally licensed to AbbVie Inc. (AbbVie) (see AbbVie below), the Company’s collaboration revenue and deferred revenue have been generated primarily from licensing fees and reimbursements for expenses received under our exclusive license with Kyowa Kirin (the Kyowa Kirin Agreement).

Kyowa Kirin

In December 2009, the Company entered into an exclusive license with KKC (the KKC Agreement)Kyowa Kirin to develop and commercialize bardoxolone in the licensed territory. The terms of the agreement include payment to the Company of a nonrefundable, up-front license fee of $35.0$35.0 million and additional development and commercial milestone payments. As of March 31, 2021,2022, the Company has received $45.0$50.0 million related to regulatory development milestone payments from KKCKyowa Kirin and has the potential in the future to achieve another $52.0$47.0 million from 6 regulatory milestones and $140.0$140.0 million from 4 commercial milestones. The Company also has the potential to achieve tiered royalties ranging from the low teens to the low 20 percent range, depending on the country of sale and the amount of annual net sales, on net sales by KKCKyowa Kirin in the licensed territory. The Company is participating on a joint steering committee with KKCKyowa Kirin to oversee the development and commercialization activities related to bardoxolone. Any future milestones and royalties received are subject to mid to lower single digit percent declining tiered commissions to certain consultants as compensation for negotiations of the KKCKyowa Kirin Agreement.

The up-front payment and regulatory milestones are accounted for as a single unit of accounting. The Company regularly evaluates its remaining performance obligation under the KKCKyowa Kirin Agreement. Accordingly, revenue may fluctuate from period to period due to changes to its estimated performance obligation period and variable considerations. The Company began recognizing revenue related to the up-front payment upon execution of the KKCKyowa Kirin Agreement.

In March 2021, the Company'sCompany’s performance obligation period under the KKCKyowa Kirin Agreement was extended to June 2022 , which decreased quarterly revenue recognition by approximately $0.4$0.4 million for the three months ended March 31,prospectively.

On July 27, 2021, comparedKyowa Kirin submitted a New Drug Application (NDA) in Japan to the three months ended March 31, 2020.  TheMinistry of Health, Labour and Welfare for bardoxolone for improvement of renal function in patients with Alport syndrome. Based on this submission, the Company recognized collaboration revenue totaling approximately $0.8 and $1.2earned a $5.0 million during each ofmilestone payment, variable consideration previously considered constrained, under the three months ended March 31, 2021 and 2020, respectively.Kyowa Kirin Agreement. As of March 31, 2021,a result, the Company recorded $4.7 million in collaboration revenue, a cumulative catch-up for the portion of this milestone that was satisfied in prior periods, and $0.3 million in deferred revenue totaling approximately $3.9 million, which is reflected asthat will be recognized over the current portion of deferred revenue.remaining performance obligation period, ending in June 2022.

AbbVie

In September 2010, the Company entered into a license agreement with AbbVie Inc. (AbbVie) (the AbbVie License Agreement) for an exclusive license to develop and commercialize bardoxolone in the Licensee Territory (as defined in the AbbVie License Agreement).

In December 2011, the Company entered into a collaboration agreement with AbbVie (the Collaboration Agreement) to jointly research, develop, and commercialize the Company’s portfolio of second and later generation oral Nrf2 activators.

In October 2019, the Company and AbbVie entered into an Amended and Restated License Agreement (the Reacquisition Agreement) pursuant to which the Company reacquired the development, manufacturing, and commercialization rights concerning its proprietary Nrf2 activator product platform originally licensed to AbbVie in the AbbVie License Agreement and the Collaboration Agreement.Agreement. In exchange for such rights, the Company agreed to pay AbbVie $330.0$330.0 million, all of which total payments of $250.0 million havehas subsequently been made as of March 31, 2021, with the remaining $80.0 million payable on November 30, 2021.paid. Additionally, the Company will pay AbbVie an escalating, low single-digit royalty on worldwide net sales, on a product-by-product basis, of omaveloxolone and certain next-generation Nrf2 activators.

The execution of the Reacquisition Agreement ended our performance obligations under the Collaboration Agreement and included the write off of the remaining related deferred revenue balance, after which no further revenue was recognized.  Accordingly, there was 0 revenue recognized in 2020.  Agreement.


9


The Company recognized interest expense related to the Reacquisition Agreement of approximately $1.7 million and $1.6$1.7 million, during the three months ended March 31, 2021 and 2020, respectively.2021. As of March 31, 2021, 2022, the Company’sCompany has fully satisfied its payable to collaborators was $80.0 million, with a present value of $75.2 millionAbbVie, therefore .  0

4. Term Loan

On October 9, 2019, the Company entered into the First Amendment to the Amended and Restated Loan and Security Agreement (the Amended Restated Loan Agreement), under which it borrowed $155.0 million as of December 20, 2019.  On June 24, 2020, the Company paid off the total outstanding balance of the term loans under the Amended Restated Loan Agreement (Term Loans) prior to the maturity date.  The payoff consisted of (i) the outstanding principal balance of $155.0 million, (ii) exit fees of $6.7 million, which has been partially accrued up to the date of repayment, (iii) prepayment fees of $5.4 million, and (iv) accrued and unpaid interest of $1.0 million.  At the time of payoff, all liabilities and obligations under the Amended Restated Loan Agreement were terminated. The Company recognized approximately $0 and $5.9 million in interest expense was recognized for the three months ended March 31, 2021 and 2020, respectively.2022.

5.4. Liability Related to Sale of Future Royalties

On June 24, 2020, the Company closed on the Development and Commercialization Funding Agreement with an affiliate of Blackstone Life Sciences, LLC (BXLS), which provides funding for the development and commercialization of bardoxolone for the treatment of CKD caused by Alport syndrome, autosomal dominant polycystic kidney disease (ADPKD), and certain other rare CKD indications in return for future royalties (the Development Agreement). The Development Agreement includes a $300.0$300.0 million payment by an affiliate of BXLS in return for various percentage royalty payments on worldwide net sales of bardoxolone, once approved in the United States or certain specified European countries, by Reata and its licensees, other than KKC.Kyowa Kirin. The royalty percentage will initially be in the mid-single digits and, in future years, can vary between higher-mid single digit percentages to low-single digit percentages depending on various milestones, including indication approval dates, cumulative royalty payments, and cumulative net sales. Pursuant to the Development Agreement, we have granted BXLS a security interest in substantially all of our assets.After a bardoxolone product approval has been obtained by the Company, the Company is obligated to make certain minimum cumulative payment amounts in 2025 through 2033, but only until BXLS has achieved certain internal rate of return target.

In addition, concurrent with the Development Agreement, the Company entered into a common stock purchase agreement (the Purchase Agreement) with affiliates of BXLS to sell an aggregate of 340,793 shares of the Company’s Class A common stock at $146.72$146.72 per share for a total of $50.0$50.0 million.

The Company concluded that there were 2 units of accounting for the consideration received, comprised of the liability related to the sale of future royalties and the common shares. The Company allocated the $300.0$300.0 million from the Development Agreement and $50.0$50.0 million from the Purchase Agreement between the two units of accounting on a relative fair value basis at the time of the transaction. The Company allocated $294.5$294.5 million, which includes $0.8$0.8 million in transaction costs incurred, in transaction consideration to the liability, and $55.5$55.5 million to the common shares. The Company determined the fair value of the common shares based on the closing stock price on the June 24, 2020, the closing date of the Development Agreement. The effective interest rate under the Development Agreement, including transaction costs, is approximately 13.8%13.8%. The Company reassessed the expected royalty payments and lowered our previous estimate of future sales for which royalties will be paid. Accordingly, we have prospectively adjusted and recognized lower non-cash interest expense using a 10.9% effective interest rate, as of March 31, 2022.

The following table shows the activity within the liability related to sale of future royalties for the three months ended March 31, 2021:2022:

 

Liability Related to Sale of Future Royalties

 

 

(in thousands)

 

Balance at December 31, 2021

$

362,928

 

Non-cash interest expense recognized

 

9,855

 

Balance at March 31, 2022

 

372,783

 

Less: Unamortized transaction cost

 

(770

)

Carrying value at March 31, 2022

$

372,013

 

10


 

Liability Related to Sale of Future Royalties

 

 

(in thousands)

 

Balance at December 31, 2020

$

316,305

 

Non-cash interest expense recognized, net of transaction cost amortization

 

10,909

 

Balance at March 31, 2021

 

327,214

 

Less: Unamortized transaction cost

 

(835

)

Carrying value at March 31, 2021

$

326,379

 


6.5. Other Income (Expense), Net

 

 

Three Months Ended

 

 

Three Months Ended

 

 

March 31

 

 

March 31

 

 

2022

 

 

2021

 

 

2021

 

 

2020

 

 

(in thousands)

 

Other income (expense), net

 

 

 

 

 

 

 

 

 

 

 

 

 

Investment income

 

$

80

 

 

$

2,055

 

 

$

132

 

$

80

 

Interest expense

 

 

(1,714

)

 

 

(5,869

)

 

0

 

(1,714

)

Non-cash interest expense on liability

related to sale of future royalty

 

 

(10,925

)

 

 

0

 

 

(9,871

)

 

(10,925

)

Other income (expense)

 

 

3

 

 

 

0

 

 

 

(33

)

 

 

3

 

Total other income (expense), net

 

$

(12,556

)

 

$

(3,814

)

 

$

(9,772

)

 

$

(12,556

)

Investment Income

Interest income consists primarily of interest generated from our cash and cash equivalents.

Interest Expense

Interest expense consists primarily of interest on our borrowing activities under our loan agreements and the imputed interest from amount due to AbbVie under the Reacquisition Agreement.

Non-Cash Interest Expense on Liability Related to Sale of Future Royalties

Non-cash interest expense consists of recognition of interest expense based on the Company’s current estimate of future royalties expensed to be paid over the estimated term of the Development Agreement.

Other Income (Expense)

Other income (expense) consists primarily of gains and losses on foreign currency exchange.

6. Leases

7. Leases

The Company’sCompany headquarters areis located in Plano, Texas, where it leases approximately 122,000 square feet of office space. On September 2, 2020, the Company’s sublease agreement for its offices in Plano, Texas, was terminated due to the bankruptcy filing of its lessor. On October 1, 2020, the Company entered into a lease agreement with the owner of its offices in Plano, Texas, with lease terms extending through June 30, 2022 and an option to renew up to three months.

The Company leases additional space located in Irving, Texas, where it leases approximately 34,890 square feet of office and laboratory space.

On February 4, 2022, the Company extended the lease for the office and laboratory space of approximately 34,890 square feet located in Irving, Texas, with lease terms extending through April 30, 2022to October 31, 2024, with an option to renew up to four successive extendthree-month for a fixed periods.   twelve-month period.

TheOn March 8, 2022, the Company has elected to net the amortization of the right-of-use assets and the reduction ofextended the lease liabilities principal in accrued direct research and other current and long-term liabilities in the consolidated statements of cash flows. During the three months ended March 31, 2021, cash paid for amounts included for the measurement of lease liabilities was $0.8 million. During the three months ended MarchPlano office to December 31, 2021, the Company recorded operating lease expense of $0.8 million.  2023.

Supplemental balance sheet information related to the Company’s operating leases is as follows:


 

 

 

 

As of March 31,

 

 

 

Balance Sheet Classification

 

2021

 

 

2020

 

 

 

 

 

(in thousands, except for years and %)

 

Non-current right-of-use assets

 

Other assets

 

$

4,509

 

 

$

8,369

 

Current lease liabilities

 

Other current liabilities

 

$

2,905

 

 

$

3,367

 

Non-current lease liabilities

 

Other long-term liabilities

 

$

1,730

 

 

$

6,107

 

 

 

 

 

 

 

 

 

 

 

 

Weighted-average remaining lease term (in years)

 

 

 

 

1.5

 

 

 

2.6

 

Weighted-average discount rate

 

 

 

 

8.1

%

 

 

9.6

%

Maturities of lease liabilities by fiscal year for the Company’s operating leases:

 

 

As of March 31, 2021

 

 

 

(in thousands)

 

2021 (remaining nine months)

 

$

2,376

 

2022

 

 

2,570

 

Total lease payments

 

 

4,946

 

Less: Imputed interest

 

 

(311

)

Present value of lease liabilities

 

$

4,635

 

The Company has an additional lease of a single-tenant, build-to-suit building of approximately 327,400 square feet of office and laboratory space located in Plano, Texas with an initial lease term of 16 years. The Company entered into the lease agreement on October 15, 2019 (the 2019 Lease Agreement), and at the Company’s option, it may renew the lease for two consecutive five-year renewal periods or one ten-year renewal period.  Theperiod. On December 15, 2021, the Company does not haveobtained control of the space, orand, accordingly, the construction prior to completion of construction.  Therefore, 0Company recorded related right-of-use orassets and the lease liabilities wereduring the fourth quarter of 2021. The Company recorded in connectionthe liability associated with the 2019 Lease Agreement at the present value of the lease payments not yet paid, using the discount rate as of March 31, 2021.  Under the First Amendment tocommencement date. As the discount rate implicit in the 2019 Lease Agreement executedwas not readily determinable, the Company utilized its incremental borrowing rate. The renewals are not assumed in May 2020, the landlord will funddetermination of the Company’s leasehold improvements uplease term, since they are not deemed to $31.3be reasonably assured at the inception of the lease. At inception, the Company recorded $124.5 million as a right-of-use asset, which represented a lease liability of $133.2 million, net of $8.7 million of which the Company has recorded a leasehold incentive obligation of approximately $2.7 million as other long-term liabilities as of March 31, 2021.  The initial annual base rent will be determined based on the project cost, subject to an initial annual cap of approximately $13.3 million, which may increase in certain circumstances.  Beginning in the third lease year, the base rent will increase 1.95% per annum each year.  In addition to the annual base rent, the Company will pay for taxes, insurance, utilities, operating expenses, assessments under private covenants, maintenance and repairs, certain capital repairs and replacements, and building management fees.  incentives recognized.

8. Income Taxes11


On March 27, 2020, the United States enacted the CARES Act.  The CARES Act is an emergency economic stimulus package that includes spending and tax breaks to strengthen the U.S. economy and to provide assistance to individuals, families, and businesses affected by COVID-19.  Accordingly, under its provisions, in March 2020, the Company recognized tax benefits and receivables totaling $22.2 million associated with the ability to carryback an applicable prior year’s net operating losses to a preceding year, which had previously been fully reserved by its valuation allowance.  In April 2021, the Company received $2.9 million out of the $22.2 million of the income tax receivable as of March 31, 2021.

For the three months ended March 31, 2021,2022, the Company’s effective tax rate was 0% compared to a benefitCompany paid $0.8 million for amounts included in the measurement of 31.3% forlease liabilities. During the three months ended March 31, 2020.  2022, and 2021, the Company recorded total rent expense of $4.3 million and $0.8million, respectively.

Supplemental balance sheet and other information related to the Company’s operating leases is as follows:

 

 

 

 

As of March 31,

 

 

 

 

 

2022

 

 

2021

 

Weighted-average remaining lease term (in years)

 

 

15.6

 

 

 

1.5

 

Weighted-average discount rate

 

 

 

 

6.5

%

 

 

8.1

%

Maturities of lease liabilities by fiscal year for the Company’s operating leases:

 

 

As of March 31, 2022

 

 

 

(in thousands)

 

2022 (remaining nine months)

 

$

9,748

 

2023 (1)

 

 

10,638

 

2024

 

 

7,427

 

2025

 

 

13,737

 

Thereafter

 

 

196,049

 

Total lease payments (1)

 

 

237,599

 

Less: Imputed interest

 

 

(96,012

)

Present value of lease liabilities

 

$

141,587

 

(1) Above table assumes one year rent abatement is applied beginning in June 2023 following United States Food and Drug Administration (FDA) approval of omaveloxolone.

7. Income Taxes

The following table summarizes income tax (benefit) expense and effective income tax rate:

 

 

Three Months Ended

 

 

 

March 31

 

 

 

2022

 

 

2021

 

 

 

(in thousands, except for percentage data)

 

Benefit from (provision for) taxes on income

 

$

(31

)

 

$

15

 

Effective income tax rate

 

 

0.0

%

 

 

0.0

%

The Company’s effective tax rate for the three months ended March 31, 20212022, varies with the statutory rate primarily due to changes in the valuation allowance related to certain deferred tax assets generated or utilized in the applicable period.

Deferred tax assets are regularly reviewed for recoverability by jurisdiction and valuation allowances are established based on historical and projected future taxable losses and the expected timing of the reversals of existing temporary differences. The


Company has recorded valuation allowances against the majority of its deferred tax assets as of March 31, 2021,2022, and the Company expects to maintain these valuation allowances until there is sufficient evidence that future earnings can be achieved, which is uncertain at this time.

9.12


8. Stock-Based Compensation

The following table summarizes time-based and performance-based stock compensation expense reflected in the consolidated statements of operations (in thousands):operations:

 

Three Months Ended

 

 

Three Months Ended

 

 

March 31

 

 

March 31

 

 

2022

 

 

2021

 

 

2021

 

 

2020

 

 

(in thousands)

 

Research and development

 

$

6,808

 

 

$

11,516

 

 

$

7,606

 

$

6,808

 

General and administrative

 

 

7,871

 

 

 

7,791

 

 

 

7,838

 

 

 

7,871

 

Total stock compensation expense

 

$

14,679

 

 

$

19,307

 

 

$

15,444

 

 

$

14,679

 

Restricted Stock Units (RSUs)

The following table summarizes RSU activity as of March 31, 2021,2022, under the Second Amended and Restated Long Term Incentive Plan (LTIP Plan) agreement:

 

Number of

RSUs

 

 

Weighted-Average

Grant Date Fair

Value

 

Outstanding at January 1, 2021

 

 

108,551

 

 

$

115.54

 

 

Number of
RSUs

 

 

Weighted-Average
Grant Date Fair
Value

 

Outstanding at January 1, 2022

 

809,145

 

$

66.91

 

Granted

 

 

260,408

 

 

 

121.46

 

 

513,559

 

27.36

 

Vested

 

 

(3,302

)

 

 

212.12

 

 

(37,942

)

 

110.88

 

Forfeited

 

 

(12,199

)

 

 

128.19

 

 

 

(67,917

)

 

71.76

 

Outstanding at March 31, 2021

 

 

353,458

 

 

$

118.56

 

Outstanding at March 31, 2022

 

 

1,216,845

 

 

$

48.58

 

As of March 31, 2021,2022, total unrecognized compensation expense related to RSU and performance-based RSUs awards that were deemed probable of vesting was approximately $8.1$40.8 million, which excludes 291,072148,000 shares of unvested performance-based RSUs that were deemed not probable of vesting totaling unrecognized stock-based compensation expense of $33.1$14.2 million.

Stock Options

The following table summarizes stock option activity as of March 31, 2021,2022, under the LTIP Plan and standalone option agreements:

 

 

Number of

Options

 

 

Weighted-

Average

Price

 

Outstanding at January 1, 2021

 

 

4,306,269

 

 

$

79.47

 

Granted

 

 

697,538

 

 

 

123.39

 

Exercised

 

 

(112,423

)

 

 

41.59

 

Forfeited

 

 

(94,271

)

 

 

138.30

 

Expired

 

 

(30,727

)

 

 

205.84

 

Outstanding at March 31, 2021

 

 

4,766,386

 

 

$

84.81

 

Exercisable at March 31, 2021

 

 

2,282,312

 

 

$

46.62

 

Stock-based compensation expense for the three months ended March 31, 2021, included accelerated recognition of expense of $1.4 million due to modifications of outstanding stock options as a result of employees


 

 

Number of
Options

 

 

Weighted-
Average
Price

 

Outstanding at January 1, 2022

 

 

4,743,180

 

 

$

86.06

 

Granted

 

 

1,110,981

 

 

 

27.78

 

Exercised

 

 

(9,375

)

 

 

21.25

 

Forfeited

 

 

(175,492

)

 

 

114.33

 

Expired

 

 

(7,468

)

 

 

145.19

 

Outstanding at March 31, 2022

 

 

5,661,826

 

 

$

73.78

 

Exercisable at March 31, 2022

 

 

3,013,168

 

 

$

59.05

 

who entered into consulting agreements at the termination of employment, which were considered to be non-substantive services.

As of March 31, 2021,2022, total unrecognized compensation expense related to stock options was approximately $125.5$88.4 million, which excludes 482,800568,450 shares of unvested performance-based stock options that were deemed not probable of vesting totaling unrecognized stock-based compensation expense of $48.8$49.4 million.

The total intrinsic value of all outstanding options and exercisable options atas of March 31, 20212022 was $183.7$24.5 million and $139.6$18.9 million, respectively.

10. 13


The number of weighted average options that were not included in the diluted earnings per share calculation because the effect would have been anti-dilutive represented 6,878,671 and 4,766,386 shares as of March 31, 2022 and 2021, respectively.

9. Employee Benefit Plans

In 2010, we adopted an Employee Investment Plan, qualified under Section 401(k) of the Internal Revenue Code, which is a retirement savings plan covering substantially all of our U.S. employees (the Plan). The Plan is administered under the “safe harbor” provision of ERISA. Under the Plan, an eligible employee may elect to contribute a percentage of their salary on a pre-tax basis, subject to federal statutory limitations. Beginning in January 2019, the Company implemented a discretionary employer matching contribution of $1.00$1.00 for every $1.00$1.00 contributed by a participating employee up to $5,000$7,000 and $6,000 annually in 2022 and 2021, respectively, which such matching contributions become fully vested after four years of service. The Company recorded expense of $0.8$1.3 million and $0.6$0.8 million for the three months ended March 31, 20212022 and 2020,2021, respectively, which includes the Company’s contributions and administrative costs.

11. 10. Commitments and Contingencies

Litigation

From time to time, the Company is a party to legal proceedings in the course of its business, including the matters described below. The outcome of any such legal proceedings, regardless of the merits, is inherently uncertain. In addition, litigation and related matters are costly and may divert the attention of our management and other resources that would otherwise be engaged in other activities. If the Company were unable to prevail in any such legal proceedings, its business, results of operations, liquidity and financial condition could be adversely affected. The Company recognizes accruals for litigations to the extent that it can conclude that a loss is both probable and reasonably estimable and recognizes legal expenses as incurred.

PatelBardoxolone Securities Litigation

On October 15, 2020, Toshif PatelFour putative stockholders of the Company filed a complaintcomplaints for alleged violationviolations of the federal securities laws against the Company and certain of its executives, including its Chief Executive Officer, its Chief Operating Officer and Chief Financial Officer, and its Chief FinancialInnovation Officer (in one of the suits), three of the complaints were filed in the United States District Court for the Eastern District of Texas.Texas, and one was filed in the District of New Jersey. The complaint purports to bring a federal securities class action on behalf of a class of persons who acquired the Company’s common stock between October 15, 2019 and August 7, 2020.  The complaint alleges,complaints allege, among other things, that the defendantsCompany (and certain of its executives) made false and misleading statements regarding the sufficiency of its MOXIe Part 2the Phase 3 CARDINAL study results to support a single study marketing approval of omaveloxolonean NDA for bardoxolone in the treatment of FACKD caused by Alport syndrome, and the Company’s interactions with the FDA concerning the study. The complaints filed in the United States.States District Court for the Eastern District of Texas, were consolidated on April 22, 2022, and the complaint filed in the District of New Jersey on February 18, 2022, which subsequently was voluntarily dismissed. The plaintiffs seek, among other things, a class action designation, an award of damages, and costs and expenses, including attorney fees and expert fees. The Company currently expects a single, consolidated, amended complaint to be filed in the future.

The Company believes that the allegations contained in the complaints are without merit and intends to defend the cases. The Company cannot predict at this point the length of time that these actions will be ongoing or the liability, if any, which may arise therefrom.

Derivative Lawsuit

An alleged stockholder of the Company filed a derivative action in the Court of Chancery of the State of Delaware against all of the directors of the Company and naming the Company as a nominal defendant. The plaintiff asserts claims in the complaint of breach of fiduciary duty and unjust enrichment concerning the alleged payment of excessive compensation to the non-employee directors of the Company between fiscal years 2019 and 2021. The plaintiff seeks, among other things, the designation of this action as a class action, an award of unspecified compensatoryorder awarding damages and interest, costs and expenses, including counselattorneys and expert fees, and expert fees.directing the Board of Directors to reform and improve its corporate governance and internal procedures relating to the award of non-employee director compensation.

14


The Company believesdefendants believe that the allegations contained in the complaint are without merit and intendsintend to defend the case vigorously.case. The Company cannot predict at this point the length of time that this action will be ongoing or the liability, if any, which may arise therefrom.

Indemnifications

ASCAccounting Standards Codification 460, Guarantees, requires that, upon issuance of a guarantee, the guarantor must recognize a liability for the fair value of the obligations it assumes under that guarantee.


As permitted under Delaware law and in accordance with the Company’s bylaws, officers and directors are indemnified for certain events or occurrences, subject to certain limits, while the officer or director is or was serving in such capacity. The maximum amount of potential future indemnification is unlimited; however, the Company has obtained director and officer insurance that limits its exposure and may enable recoverability of a portion of any future amounts paid. The Company believes the fair value for these indemnification obligations is minimal. Accordingly, the Company has not recognized any liabilities relating to these obligations as of March 31, 2021.  2022.

The Company has certain agreements with licensors, licensees, collaborators, and collaboratorsvendors that contain indemnification provisions. In such provisions, the Company typically agrees to indemnify the licensor, licensee, collaborator, or collaboratorvendor against certain types of third-party claims. The Company accrues for known indemnification issues when a loss is probable and can be reasonably estimated. There were 0 accruals for expenses related to indemnification issues for any period presented.

11. Subsequent Events

12. Net Loss per Share

The following table sets forthOn May 6, 2022, an alleged stockholder of the computation of basic and diluted net loss per share attributable to common stockholders:

 

 

Three Months Ended

 

 

 

March 31

 

 

 

2021

 

 

2020

 

Numerator

 

 

 

 

 

 

 

 

Net loss

 

$

(67,455

)

 

$

(48,939

)

Denominator

 

 

 

 

 

 

 

 

Weighted-average number of common shares

   used in net loss per share—basic

 

 

36,203,631

 

 

 

33,222,085

 

Dilutive potential common shares

 

 

0

 

 

 

0

 

Weighted-average number of common shares

   used in net loss per share—diluted

 

 

36,203,631

 

 

 

33,222,085

 

Net loss per share – basic

 

$

(1.86

)

 

$

(1.47

)

Net loss per share – diluted

 

$

(1.86

)

 

$

(1.47

)

The number of weighted average options that were not includedCompany filed a derivative action in the diluted earnings per share calculation becauseCourt of Chancery of the effect would have been anti-dilutive represented 4,766,386State of Delaware against all of the directors of the Company and 4,861,425 sharesnaming the Company as a nominal defendant. See Note 10, Commitments and Contingenciesof March 31, 2021 and 2020, respectively.Notes to Unaudited Consolidated Financial Statements for a description of the litigation.


15


Item 2. Management’s Discussion and Analysis of Financial Condition and Results of Operations

You should read the following discussion and analysis of our financial condition and results of operations together with our consolidated financial statements and related notes and other financial information appearing in this Quarterly Report on Form 10-Q. Some of the information contained in this discussion and analysis or set forth elsewhere in this Quarterly Report on Form 10-Q, including information with respect to our plans and strategy for our business, operations, and product candidates, includes forward-looking statements that involve risks and uncertainties. Factors that may cause actual results to differ materially from current expectations include, among other things, those described under the headings “Risk Factors” and “Cautionary Note Regarding Forward-Looking Statements” and discussed elsewhere in this Quarterly Report on Form 10-Q.

Overview

We are a clinical-stage biopharmaceutical company focused on identifying, developing, and commercializing innovative therapies that change patients’ lives for the better. We concentrate on small-molecule therapeutics with novel mechanisms of action for the treatment of severe, life-threatening diseases with few or no approved therapies. Our lead programs are omaveloxolone in FA and bardoxolone in rare forms of CKD and a rare neurological disease.  We announced positive topline data from registrational trials for bothCKD. Both of our lead product candidates bardoxolone in patients with CKD caused by Alport syndrome and omaveloxolone in patients with a neurological disorder called Friedreich’s ataxia (FA).  Both bardoxolone and omaveloxolone activate the transcription factor Nrf2 to normalize mitochondrial function, restore redox balance, and resolve inflammation. Because mitochondrial dysfunction, oxidative stress, and inflammation are features of many diseases, we believe omaveloxolone, bardoxolone, and omaveloxoloneour next-generation Nrf2 activators have many potential clinical applications. We possess exclusive, worldwide rights to develop, manufacture, and commercialize omaveloxolone, bardoxolone, omaveloxolone, and our next-generation Nrf2 activators, excluding certain Asian markets for bardoxolone in certain indications, which are licensed to KKC.Kyowa Kirin. In addition, we are developing RTA 901, the lead product candidate from our Hsp90 modulator program, in neurological indications. We are the exclusive licensee of RTA 901 and have worldwide commercial rights.

Recent Key Developments

Omaveloxolone for Friedreich’s Ataxia

The FDA has granted Fast Track Designation, Orphan Drug Designation, and Rare Pediatric Disease Designation to omaveloxolone for the treatment of FA. In March 2022, we completed rolling submission of an NDA to the FDA for omaveloxolone for the treatment of patients with FA. This NDA is supported by the efficacy and safety data from the MOXIe Part 2 trial and additional supporting data from the MOXIe Part 1 and MOXIe Extension trials.

We are continuing to complete the regulatory procedures and submissions required prior to filing a Marketing Authorization Application (MAA) in Europe for approval of omaveloxolone for the treatment of patients with FA. We have secured agreement on our Pediatric Investigation Plan with the Pediatric Committee, and we also received European Medicines Agency (EMA) Follow-Up Protocol Assistance feedback regarding our nonclinical and chemistry manufacturing controls (CMC) programs. The EMA feedback indicated that there were no identified impediments to our planned MAA submission and included agreement that certain nonclinical studies, including 2- year carcinogenicity study data, may be submitted after approval. We plan to submit an MAA to the European Medicines Agency for omaveloxolone in the fourth quarter of 2022.

RTA 901 for Neurological Indications, Including Diabetic Peripheral Neuropathic Pain

In the first quarter of 2022 we initiated additional Phase 1 clinical pharmacology studies of RTA 901, including a drug-drug interaction study. Following completion of these Phase 1 studies, we plan to initiate a randomized, double-blind placebo-controlled Phase 2 trial of RTA 901 in diabetic patients with peripheral neuropathic pain in the fourth quarter of 2022.

Bardoxolone in Patients withCKD Caused by Alport Syndrome

On April 26, 2021, we announced thatWe received a Complete Response Letter (CRL) from the U.S. Food and Drug Administration (FDA) accepted for filing the New Drug Application (NDA)FDA in February 2022 with respect to its review of our NDA for bardoxolone forin the treatment of patients with CKD caused by Alport syndrome. The FDA will review the application under a Standard Review timeline.  The Prescription Drug User Fee Act (PDUFA) date,CRL indicated the FDA action datecannot approve the NDA in its present form. We will continue to work with the FDA to confirm our next steps on our Alport syndrome program.

16


In October 2021, we submitted an MAA to the EMA for the application, is scheduled for February 25, 2022.  The FDA also advised us that it is currently planning to hold an Advisory Committee meeting to discuss the application.

Our NDA submission was based on the results of Year 2 of the Phase 3 CARDINAL study of bardoxolone in the treatment of patients with CKD caused by Alport syndrome announced in November 2020. The study met its primary and key secondary endpoints following two yearssyndrome. In the first quarter of treatment (referred to as Year 2). Moreover,2022, we also announced that patients who completed one yearreceived the 120-day list of questions from the EMA. We are in the EAGLE long-termprocess of preparing our responses. We requested a 90-day extension study and were treated with bardoxolone for a total of three years (n=14) showed a sustained and significant increase from baseline in estimated glomerular filtration rate (eGFR).  Together, these data suggest that bardoxolone treatment has beneficial long-term effects on kidney function in patients with Alport syndrome.

We also plan to pursue marketing approval outside ofour responses which was granted by the United States. We plan to submit a Marketing Authorization Application (MAA) with the European Medicines Agency (EMA) in the fourth quarter of 2021 for marketing approval of bardoxoloneEMA. The timeline for the treatment of CKD caused by Alport syndrome in the European Union.EMA’s review cycle was therefore extended.

Bardoxolone in Patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD)

We are currently enrolling patients in FALCON, ana Phase 3, international, multi-center, randomized, double-blind, placebo-controlled trial studying the safety and efficacy of bardoxolone in patients with ADPKD, randomized one-to-one to active drug or placebo. FALCON will enroll approximately 550is enrolling patients in a broad range of ages 18 to 70 years old, with an eGFRestimated glomerular filtration rate (eGFR) between 30 toand 90 mL/min/1.73 m2. The primary endpoint is the off-treatment eGFR change from baseline versus placebo at Week 52, which represents 48 weeks of treatment followed by a four-


week withdrawal period. MoreAt 52 weeks, patients resume treatment for a second 48-week period to Week 100 followed by a second four-week withdrawal period to Week 104. than 550The key secondary endpoint is the off-treatment eGFR change from baseline versus placebo at Week 104.

The FDA has provided us with guidance that, in patients with ADPKD, an analysis of eGFR during the off-treatment period demonstrating an improvement versus placebo after one year of bardoxolone treatment may support an NDA submission for accelerated approval of bardoxolone for the treatment of ADPKD, and data demonstrating an improvement versus placebo after two years of treatment may support full approval.

In March 2020, we announced a temporary pause in enrollment in FALCON due to the COVID-19 pandemic; we resumed enrollment during the third quarter of 2020. Despite the pandemic, most sites are currently able to screen and randomize patients. More than 290 patients are currently enrolled in the study, andtrial.

In the first quarter of 2022, we expectsubmitted a protocol amendment to complete enrollment inthe FALCON by the end of 2021.

Bardoxolone in Patients with CKD at Risk of Rapid Progression

MERLIN is a multi-center, double-blind, placebo-controlled, Phase 23 trial to evaluate the safety and efficacy of bardoxolone in patients at riskwith ADPKD with the FDA and other relevant health authorities. As agreedwith the FDA prior to submission of rapidly progressing CKD duethe amendment, we recently had a Type A meeting to multiple etiologies including IgA Nephropathy (IgAN), focal segmental glomerulosclerosis (FSGS), type 1 diabetic CKD (T1D CKD), type 2 diabetic CKD (T2D CKD), hypertensive CKD,discuss key changes made to the FALCON protocol. Based on the discussion during the meeting and others. Thethe meeting minutes, the Division stated that the proposed primary endpoint of the trial iseGFR change in eGFR from baseline toat Week 12.  We expect to complete enrollment108 (8 weeks after planned drug discontinuation at Week 100) was reasonable since the available data suggest that bardoxolone’s acute pharmacodynamic effect on eGFR should be largely resolved. The Division provided guidance on handling of data from patients who completed Year 2 of the study before the protocol amendment and so did not have an eGFR assessment at Week 108. This included direction on imputing missing data for these patients in the MERLIN trial byprimary analysis. The Division stated that, in addition to the endprimary endpoint, it will be important to demonstrate that the treatment effect accrues over time to support a claim that bardoxolone slows the loss of the second quarter of 2021, and data are expected in the second half of 2021. If the results of this study are positive, we would potentially proceed to a larger Phase 3 trial with similar eligibility criteria. Patients at risk for rapid progression experience a significant risk of progressing to end-stage kidney disease (ESKD) and are a population with high unmet need across multiple forms of CKD.

Omaveloxolone in Patients with Friedreich’s Ataxia (FA)

Data from the registrational Part 2 portion of the MOXIe Phase 2 trial of omaveloxolonefunction in patients with FA (MOXIe Part 2)ADPKD and provided guidance on the open-label extension study (the MOXIe Extension) were analyzed in additional exploratory analyses (the Delayed-Start Analyses), whereby parallel trajectories between the patients randomized to placebo (placebo-to-omaveloxolone group) and those randomized to omaveloxolone (omaveloxolone-to-omaveloxolone group) in the double-blind period from MOXIe Part 2, through 48 weeks in the MOXIe Extension could provide evidence of disease-modifying activity.  A total of 73 out of 75 (97%) patients without pes cavus who completed MOXIe Part 2 enrolled in the MOXIe Extension. A longitudinal analysis used to calculate annualized slopes incorporating all available data from the MOXIe Extension showed similar slopes in Modified Friedreich’s Ataxia Rating Scale (mFARS)statistical methodologies for the placebo-to-omaveloxolone group (0.59 points per year) when compared to the omaveloxolone-to-omaveloxolone group (0.41 points per year) with no significant difference between slopes (p=0.75).exploratory eGFR slope analyses. The resulting parallel trajectories between both treatment groups are consistent with disease-modifying activity.FDA also confirmed that if FALCON is positive, it could support registration of bardoxolone in ADPKD.

We believe that the results of the Delayed-Start Analyses suggest disease-modifying activity with omaveloxolone, provide evidence supporting the positive primary endpoint findings in MOXIe Part 2, and provide additional evidence of the effectiveness of omaveloxolone in patients with FA. We requested and the FDA has granted us a Type C meeting, which is scheduled to occur in the second quarter of 2021, to discuss the Delayed-Start Analyses and the FA development program.  We plan to initiate a second pivotal study of omaveloxolone in FA patients in the fourth quarter of 2021, incorporating input from both the FDA and the EMA into the protocol before we initiate enrollment.

Update on Adjustments to Operations Due to COVID-19

In accordance with recommendations from local, state, and national authorities, our offices were opened, with additional safety protocolsWhen COVID-19 emerged as a global pandemic in place, to select groups of certain employees during the first quarter of 2021.  We are currently developing plans for2020, Reata was quick to respond and was an early adopter of a phased approachwork-from-home policy, with the exception of the laboratory that continued to reopening our officesoperate throughout under strict safety protocols. For all remote employees, we provided appropriate workstation equipment as well as training and resources to all employees.support employees’ mental and emotional wellbeing. In the second quarter of 2021, Reata relaxed its policy and permitted employees to return to the office as required. Although the Omicron variant temporarily caused us to again restrict office attendance, in February 2022 we permitted modified work schedules to meet business and personal needs.


Background: Our Programs

The following chart outlines each of our programs by indication and phase of development:

img70967532_0.jpg 

In addition, KKC,1Rolling NDA submission completed in March 2022.

2DPNP: Diabetic peripheral neuropathic pain.

17


3On February 25, 2022, we received a CRL from the FDA. We will continue to work with the FDA to confirm our next steps on our Alport syndrome program. MAA in EU is under review.

4AYAME trial conducted in Japan by our strategic collaborator in CKD, is currently conducting its registrational trialKyowa Kirin. Kyowa Kirin expects the last patient visit in the second half of 2022.

5Based on the outcome of AYAME and FALCON trials, and our discussions with the FDA regarding the bardoxolone program, we will decide future development plans for bardoxolone in additional forms of CKD.

Programs in Neurological Diseases

We are developing omaveloxolone for the treatment of patients with FA, an inherited, debilitating, and degenerative neuromuscular disorder that is usually diagnosed during adolescence and can ultimately lead to premature death. In March 2022, we completed rolling submission of our NDA for omaveloxolone for the treatment of patients with FA. Because mitochondrial dysfunction is a key feature of many neuromuscular diseases, we believe omaveloxolone may be broadly applicable to treat neurological diseases by activating Nrf2 to normalize and improve mitochondrial function and adenosine triphosphate (ATP) production. We plan to pursue the development of omaveloxolone and our other Nrf2 activators for one or more additional neurological diseases.

We are also developing RTA 901 for the treatment of neurological diseases. RTA 901 is a highly potent and selective C-terminal modulator of Hsp90, which has a critical role in mitochondrial function, protein folding, and inflammation. RTA 901 has demonstrated profound efficacy in a wide range of animal models of neurological disease, including diabetic (type 1neuropathy, neuroinflammation, and 2) CKD in Japan.  KKC completed patient enrollment in thisneuropathic pain. We plan to initiate a randomized, placebo-controlled Phase 2 trial in June 2019 and expects to have topline dataDPNP in the first halffourth quarter of 2022.

*NDA accepted for filing on April 26, 2021.

**See discussion below under “OmaveloxoloneOmaveloxolone in Patients with FA”.Friedreich’s Ataxia

Patients with FA experience progressive loss of coordination, muscle weakness, and fatigue, which commonly progress to motor incapacitation and wheelchair reliance. Based on literature and proprietary research, we believe FA affects approximately 5,000 children and adults in the United States and 22,000 individuals globally. According to data provided by IQVIA in 2020, there are approximately 4,000 projected patients diagnosed with FA in the United States. The FDA has granted Orphan Drug Designation, Fast Track Designation, and Rare Pediatric Disease Designation to omaveloxolone for the treatment of FA. The European Commission has granted Orphan Drug Designation in Europe to omaveloxolone for the treatment of FA.

Diagnosis of FA typically occurs by genetic testing, and most people in the United States with FA are diagnosed in their teens and early twenties. Patients with FA experience progressive loss of coordination, muscle weakness, and fatigue that commonly results in motor incapacitation, with patients requiring a wheelchair in their twenties. The mean age of death for patients with FA is in the mid-thirties. Childhood-onset FA can occur as early as age five, is more common than later-onset FA, and normally involves more rapid disease progression. Currently, there are no approved therapies for the treatment of FA.

MOXIe Part 2 Trial Results

Part 2 of our Phase 2 trial, called MOXIe (MOXIe Part 2), was an international, multi-center, double-blind, placebo-controlled, randomized, Registrational trial that enrolled 103 patients with FA at 11 trial sites in the United States, Europe, and Australia. MOXIe Part 2 was one of the largest global, interventional trials ever completed in FA. Patients were randomized one-to-one to omaveloxolone or placebo. MOXIe Part 2 was completed in October 2019. The primary analysis population excluded patients with pes cavus (n=82), a musculoskeletal foot deformity that may interfere with the patient’s ability to perform some components of the neurological exam used to score the primary endpoint of the trial. Safety analyses were evaluated in the all-randomized population (n=103).

The primary endpoint for the trial was the change in the Modified Friedreich’s Ataxia Rating Scale (mFARS) score for omaveloxolone relative to placebo after 48 weeks of treatment. Omaveloxolone treatment demonstrated statistically significant evidence of efficacy for the primary endpoint of the trial, producing a placebo-corrected -2.40 point mean improvement in mFARS (n=82; p=0.014). Patients treated with omaveloxolone experienced a mean improvement in mFARS of -1.55 points from baseline, while patients treated with placebo experienced a mean worsening in mFARS of +0.85 points from baseline. Further, the observed placebo-corrected improvements in mFARS were time-dependent, increasing over the course of treatment with the largest improvement observed after 48 weeks of treatment.

18


img70967532_1.jpg 

Additionally, all secondary endpoints either favored the omaveloxolone arm or were neutral. Patients on omaveloxolone experienced a nominal improvement in the Activities of Daily Living (ADL) questionnaire, with all nine questions favoring the omaveloxolone arm. On average, ADL scores for patients on omaveloxolone did not change from baseline, while placebo-treated patients worsened. Both patient global impression of change (PGIC) and clinical global impression of change numerically favored omaveloxolone, and improvement in PGIC correlated with the observed improvement in mFARS.

Omaveloxolone was reported to be generally well-tolerated. Four (8%) omaveloxolone patients and two (4%) placebo patients discontinued trial drug due to an adverse event (AE). The reported AEs were generally mild to moderate in intensity, and the most common AEs (i.e., reported in > 10% of omaveloxolone-treated patients) observed more frequently (>5% difference) in omaveloxolone compared to placebo were headache, nausea, increased aminotransferases, fatigue, abdominal pain, diarrhea, oropharyngeal pain, muscle spasms, back pain, and decreased appetite. Increases in aminotransferases are a pharmacological effect of omaveloxolone. In preclinical studies, omaveloxolone has been shown to increase production of aminotransferases in vitro, which we believe are related to restoration of mitochondrial function. In MOXIe Part 2, the aminotransferase increases were associated with improvements (reductions) in total bilirubin and were not associated with any evidence of liver injury.

In MOXIe Part 2, the overall rate of serious adverse events (SAEs) was low, with five patients in the omaveloxolone group and three patients in the placebo group reporting SAEs. No new safety signals were identified, and the reported SAEs were sporadic and generally expected in FA patients. In the patients who reported SAEs while receiving omaveloxolone, none led to discontinuation.

MOXIe Extension Trial

The open-label MOXIe Extension trial is ongoing, with a total of 149 patients enrolled (57 patients from MOXIe Part 1 and 92 patients from MOXIe Part 2). A total of 73 out of 75 (97%) patients without pes cavus who completed MOXIe Part 2 were enrolled in the MOXIe Extension, including 39 patients previously randomized to placebo (the placebo-to-omaveloxolone group) and 34 patients previously randomized to omaveloxolone (the omaveloxolone-to-omaveloxolone group). Due to the COVID-19 pandemic, not all patients had mFARS assessments performed at each time point.

19


Delayed-Start Analysis Results from August 2021 Data Cut-Off

The intent of the post-hoc Delayed-Start Analysis is to evaluate whether omaveloxolone has a persistent effect on FA disease course. Conceptually, this analysis evaluates whether the treatment effect that was observed in the placebo-controlled MOXIe Part 2 trial is maintained in the MOXIe Extension trial when all patients are receiving omaveloxolone. If the treatment effect is maintained between those originally randomized to placebo (the placebo-to-omaveloxolone group) versus those originally randomized to omaveloxolone (the omaveloxolone-to-omaveloxolone group), then it demonstrates evidence of a persistent effect on the course of the disease. If the treatment effect is not maintained, and the patients originally randomized to placebo are able to achieve the same absolute response and “catch up” to the patients initially randomized to omaveloxolone, the results are consistent with a symptomatic treatment that does not affect the underlying course of the disease.

Two timepoints were used in the analysis. The first timepoint was at Week 48, the final week of treatment in the placebo-controlled MOXIe Part 2 trial. The second timepoint was at Week 72 of the open-label MOXIe Extension in which all patients received omaveloxolone. A non-inferiority test was used to evaluate if the difference in mFARS between groups observed at the first timepoint was maintained or non-inferior at the second timepoint. The analysis methods, including the specified non-inferiority margin, were based on literature (Liu-Seifert, 2015a, 2015b). When comparing treatment groups using this methodology, maintaining a negative difference between treatment groups in mFARS is evidence of a persistent treatment effect.

The Delayed-Start Analysis used in clinical modules in our initial NDA rolling submission for omaveloxolone was updated as of August 2021. In this updated analysis 58 of 73 patients from MOXIe Part 2 without pes cavus who enrolled into MOXIe Extension had at least 72 weeks of exposure in MOXIe Extension, and 28 of these patients had at least 120 weeks of exposure in the Moxie Extension.

Results of this analysis demonstrated that the between-group difference in mFARS observed at the end of the placebo-controlled MOXIe Part 2 period (least squares mean difference = -2.25 ± 1.07) was preserved at MOXIe Extension Week 72 in the delayed-start period (LS mean difference = -3.51 ± 1.45). Consistent with a persistent treatment effect on disease, the upper limit of the 90% CI for the difference estimate was less than zero (-0.615), meeting the threshold for demonstrating significant evidence of non-inferiority.

Delayed-Start Analysis Primary Endpoint (Non-Inferiority Test)1

Placebo-Controlled

Week 48 (Δ1)

Delayed-Start

Week Ex. 72 (Δ2)

Difference (LS Mean ± SE)

-2.25 ± 1.07

p=0.037

-3.51 ± 1.45

p=0.016

Estimate = Δ2 – 0.5 × Δ1

-2.39 ± 1.38

Upper Limit of 1-sided

90% CI for Estimate

-0.615

1Non-Inferiority test performed using a MMRM analysis with a Toeplitz covariance structure.

The graphical representation of changes from baseline in mFARS for omaveloxolone and placebo groups shows the separation at the end of the placebo-controlled period is maintained in the open-label period at Extension Week 72 and beyond.

20


Change from Baseline in mFARS (Patients without Pes Cavus)

img70967532_2.jpg 

Many of the visits at Week 48 and Week 72 of the MOXIe Extension were scheduled during the initial peak of COVID-19 cases during Spring to Fall 2020. The mFARS assessment must be conducted in the clinic, and many in-clinic visits did not occur due to COVID-19 related travel restrictions and site closures during this period. Apart from the data at MOXIe Extension Week 48, parallel trajectories were seen in LS Mean mFARS change from baseline between the placebo-to-omaveloxolone group and the omaveloxolone-to-omaveloxolone group in MOXIe Extension.

A longitudinal analysis was also performed to calculate annualized slopes incorporating all available data from the MOXIe Extension, which showed similar mean slopes in mFARS for the placebo-to-omaveloxolone group (0.45 ± 0.38 points per year) when compared to the omaveloxolone-to-omaveloxolone group (0.27 ± 0.59 points per year) with no significant difference between slopes (difference = -0.18 ± 0.67; p=0.79). In MOXIe Extension, omaveloxolone-treated patients have been progressing at a rate that is >75% less than the approximately two points per year that patients progressed in a recent large natural history study (Patel, 2016).

Results from the Delayed-Start Analysis indicate a persistent omaveloxolone treatment effect on the disease course of FA. Patients who received omaveloxolone during the double-blind MOXIe Part 2 had a benefit that could not be achieved by patients initially randomized to placebo who began omaveloxolone one year later in MOXIe Extension. Notably, patients previously randomized to omaveloxolone in MOXIe Part 2 continued to show mean mFARS values that were similar to their original baseline after over three years of treatment.

No new safety signals have been identified in the MOXIe Extension trial.

21


Regulatory Interactions

In the third quarter of 2021, we completed our pre-NDA meeting with the FDA. The purpose of the pre-NDA meeting was to discuss the content of Reata’s planned NDA submission including the nonclinical data and CMC packages, data standard plan, and the overall content plan. In the meeting, we stated that we believed that the MOXIe data, along with the Delayed-Start Analysis, would provide sufficient clinical data to support a full approval. The FDA stated that the proposed primary and supportive efficacy data appear reasonable, though the Delayed-Start Analysis was viewed as exploratory. The FDA noted that the ability of the data to support full approval, and the adequacy of the data and the determination of which data may be supportive of efficacy, would be a matter of review. In response to our other questions about the contents of the NDA, the FDA exercised its discretion based on the seriousness of the indication and unmet medical need, subject to review, to permit us to submit the results of certain clinical pharmacology and nonclinical studies after approval. The additional studies include a thorough QT study with omaveloxolone, nonclinical metabolite toxicity studies, and six-month and two-year nonclinical carcinogenicity studies. The need for a drug-drug interaction study with a moderate CYP3A inducer will be established upon review of the adequacy of our submitted physiological based pharmacokinetic (PK) model.

On November 18, 2021, the FDA granted omaveloxolone Fast Track Designation for the treatment of FA, providing eligibility for FDA programs such as Priority Review and rolling submission of the NDA, if relevant criteria are met. The FDA granted our request for a rolling submission, and in March 2022, we completed submission of the NDA. This NDA is supported by the efficacy and safety data from the MOXIe Part 2 trial and additional supporting data from the MOXIe Part 1 and MOXIe Extension trials.

We are continuing to complete the regulatory procedures and submissions required prior to filing a MAA in Europe for approval of omaveloxolone for the treatment of patients with FA. We have secured agreement on our Pediatric Investigation Plan with the Pediatric Committee, and we also received EMA Follow-Up Protocol Assistance feedback regarding our nonclinical and CMC programs. The EMA feedback indicated that there were no identified impediments to our planned MAA submission and included agreement that certain nonclinical studies, including 2- year carcinogenicity study data, may be submitted after approval. We plan to submit an MAA to the European Medicines Agency for omaveloxolone in the fourth quarter of 2022.

Omaveloxolone in Other Neurological Indications

Omaveloxolone is a promising platform molecule. Because mitochondrial dysfunction is a key feature of many neurological and neuromuscular diseases, we believe omaveloxolone may be broadly applicable to treat such diseases by activating Nrf2 to normalize and improve mitochondrial function and ATP production.

Based on our understanding of the pathophysiology of neurological diseases characterized by mitochondrial dysfunction, inflammation, and oxidative stress, we believe omaveloxolone may be applicable to diseases such as progressive supranuclear palsy, Parkinson’s disease, frontotemporal dementia, Huntington’s disease, amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, and epilepsy. Consistent with this, we have observed promising activity of omaveloxolone and our other Nrf2 activators in preclinical models of many of these diseases.

Our Nrf2 activators reduced seizure frequency in refractory, progressive epilepsy models and restored mitochondrial function in patient biopsy samples and preclinical models of FA, ALS, familial and sporadic Parkinson’s disease, and frontotemporal dementia. In clinical trials, improvements in neuromuscular function have been observed in FA patients treated with omaveloxolone as assessed by mFARS, and improvements in mitochondrial function, as measured by reductions in blood lactate and heart rate, have been observed in patients with primary mitochondrial disease. Accordingly, we believe that omaveloxolone has the potential to treat a number of neurological and neuromuscular diseases that currently have few or no effective therapies, and we plan to pursue the development of omaveloxolone and our other Nrf2 activators for one or more of these diseases.

RTA 901 in Neurological Diseases

RTA 901 is the lead product candidate from our Hsp90 modulator program, which includes highly potent and selective C-terminal modulators of Hsp90. We have observed favorable activity of RTA 901 in a range of preclinical models of neurological disease, including models of diabetic neuropathy, neuroinflammation, and neuropathic pain.

22


Historically, other companies have explored N-terminal Hsp90 inhibitors for cancer therapeutics; however, this approach has been associated with multiple AEs including peripheral neuropathy and ocular toxicity. Binding at the C-terminus of Hsp90 leads to increased transcription of Hsp70, a cytoprotective and molecular chaperone gene, which facilitates cell survival in response to stress without the deleterious activities of N-terminal inhibition.

In preclinical rodent disease models, we observed that RTA 901 administered orally once-daily rescued existing nerve function, restored thermal and mechanical sensitivity, and improved nerve conductance velocity and mitochondrial function. These effects are dose-dependent, reversible, and Hsp70-dependent.

We completed a Phase 1 SAD/MAD trial of oral, once-daily RTA 901 in healthy adult volunteers to evaluate the safety, tolerability, and PK profile. The PK was approximately dose-proportional up to the highest doses evaluated with a half-life ranging from two to nine hours. Human exposures easily exceeded the exposures necessary for efficacy in multiple animal models. No safety or tolerability concerns were reported. In the first quarter of 2022, we initiated additional Phase 1 clinical pharmacology studies of RTA 901, including a drug-drug interaction study. Following completion of these Phase 1 studies, we plan to initiate a randomized, double-blind placebo-controlled Phase 2 trial of RTA 901 in diabetic patients with peripheral neuropathic pain in the fourth quarter of 2022. We are the exclusive licensee of RTA 901 and have worldwide commercial rights.

Programs in Chronic Kidney Disease

We and Kyowa Kirin, our strategic collaborator in CKD in Japan, are developing bardoxolone for the treatment of patients withCKD in multiple indications, including CKD caused by Alport syndrome, ADPKD, and certain other forms of CKD that, in the aggregate, affect more than 700,000 patients in the United States.type 1 and 2 diabetic CKD. CKD is characterized by a progressive worsening in glomerular filtration rate (GFR), which is the rate at which the kidney filters waste products from the blood.  When GFR drops below approximately 15 mL/min/1.73 m2, patients develop ESKD and require dialysis or a kidney transplant to survive.  Dialysis leads to a reduced quality of life and increasesblood, called the likelihood of serious and life-threatening complications.  The five-year survivalglomerular filtration rate for hemodialysis patients is approximately 42%(GFR).

eGFR is an estimate of GFR that nephrologists use to track the decline in kidney function and progression of CKD. In 11 separate CKD clinical trials, bardoxolone has been shown to improve eGFR inWhen GFR gets too low, patients with diverse etiologies of CKD.  We believe that bardoxolone treatment has the potential to delay or prevent GFR declines that cause the need fordevelop end-stage kidney disease (ESKD) and require dialysis or a kidney transplant to survive.

We received a CRL from the FDA in February 2022 with respect to its review of our NDA for bardoxolone in the treatment of patients with CKD caused by Alport syndrome. We will continue to work with the FDA to confirm our next steps on our Alport syndrome ADPKD,program. Kyowa Kirin is currently conducting its registrational AYAME trial of bardoxolone in diabetic (types 1 and other rare forms of CKD.2) CKD in Japan.

Bardoxolone in Patients with CKD Caused by Alport Syndrome

Alport syndrome is a rare, genetic form of CKD caused by mutations in the genes encoding type IV collagen, which is a major structural component of the glomerular basement membrane in the kidney. The kidneys of patients with Alport syndrome progressively lose the capacity to filter waste products out of the blood, which can lead to ESKD and the need for chronic dialysis treatment or a kidney transplant. Alport syndrome affects both children and adults.adults and can manifest as early as the first decade of life and causes average annual declines in eGFR of approximately four to five mL/min/1.73 m2. In patients with the most severe forms of the disease, approximately 50% progress to dialysis by age 25, 90% by age 40, and nearly 100% by age 60. There are currently no approved therapies to treat CKD caused by Alport syndrome.

The Alport Syndrome Foundation estimateshas estimated that Alport syndrome affects approximately 30,000 to 60,000 people in the United States. According to data provided by IQVIA in April 2020, there are approximately 14,000 projected patients diagnosed with Alport syndrome in all stages of CKD in the United States. However, recent literature suggests that a large number of patients with Alport syndrome are either undiagnosed or misdiagnosed


with other forms of CKD.To help nephrologists identify the genetic basis of various forms of CKD, including Alport syndrome, Reata and Invitae Corporation are sponsoring the KIDNEYCODE® genetic testing program.

On November 9, 2020, we announced thatreported interim results from the Phase 3 CARDINAL study met its primary and key secondary endpoints at the end of Year 2. The Phase 3 portion of CARDINAL waslong-term extension EAGLE trial. EAGLE is an international, multi-center, double-blind, placebo-controlled, randomized registrationalopen-label, extended access trial that enrolled 157evaluating the longer-term safety and tolerability of bardoxolone in patients with CKD caused by Alport syndrome at approximately 50 study siteswho participated in the United States, Europe, Japan, and Australia. Patients were randomized one-to-one to bardoxoloneCARDINAL trial or placebo.

At Week 100,patients with ADPKD who participated in the intent to treat (ITT) population, which included eGFR values for patients who either remained on or discontinued study drug, patients treated with bardoxolone had a statistically significant improvement compared to placebo in mean change from baseline in eGFR of 7.7 mL/min/1.73 m2 (p=0.0005). Patients treated with bardoxolone experienced a mean change from baseline in eGFR of -0.8 mL/min/1.73 m2, while patients treated with placebo experienced a mean change from baseline in eGFR of -8.5 mL/min/1.73 m2.

In the modified ITT (mITT) analysis, which assessed the effect of receiving treatment by excluding values after patients discontinued treatment, patients treated with bardoxolone had a statistically significant improvement compared to placebo in mean change from baseline in eGFR at Week 100 of 11.3 mL/min/1.73 m2 (p<0.0001). In the mITT analysis, patients treated with bardoxolone experienced a mean increase from baseline in eGFR of 1.7 mL/min/1.73 m2, while patients treated with placebo experienced a mean decline from baseline in eGFR of -9.6 mL/min/1.73 m2.

At Week 104 (four weeks after last dose in second year of treatment), patients in the ITT population treated with bardoxolone had a statistically significant improvement compared to placebo in mean change from baseline in eGFR of 4.3 mL/min/1.73 m2 (p=0.023). Patients treated with bardoxolone experienced a mean change from baseline in eGFR of -4.5 mL/min/1.73 m2, while patients treated with placebo experienced a mean change from baseline in eGFR of -8.8 mL/min/1.73 m2.

Bardoxolone was generally reported to be well tolerated in this study, and the safety profile was similar to that observed in prior trials. Eight patients (10%) receiving bardoxolone and 15 patients (19%) receiving placebo experienced a treatment-emergent serious adverse event (SAE). No SAEs were reported in pediatric patients treated with bardoxolone. No fluid overload SAEs or major adverse cardiac events were reported in patients treated with bardoxolone. Blood pressure, a sensitive measure of fluid status, was not significantly different between the two groups. Weight loss was more pronounced in patients with a higher body mass index, and mean decreases in weight were not observed in pediatric patients.FALCON trial. The urinary albumin-to-creatinine ratio (UACR) was not significantly different between treatment groups at Week 100 or Week 104.

The reported adverse events (AEs) were generally mild to moderate in intensity, and the most common AEs observed more frequently in patients treated with bardoxolone compared to patients treated with placebo were muscle spasms and increases in aminotransferases which are thought to be associated with the pharmacology of the drug. Muscle spasms were generally transient and were associated with reductions of creatine kinase, which is evidence of improved energy metabolism and inconsistent with muscle injury.

Additionally, on November 9, 2020, we reported results from the long-term extension EAGLE study, in which the change from baseline in eGFR was assessed for the 14 patients with Alport syndrome who were treated with bardoxolone for three years (two years in CARDINAL and one year in EAGLE), with four-week off-treatment periods occurring at Weeks 48 and 100. Bardoxolone treatment resulted in a mean on-treatment increase from baseline in eGFR of 11.5 mL/min/1.73 m2 at Year 1, 13.3 mL/min/1.73 m2 at Year 2, and 11.0 mL/min/1.73 m2 at Year 3.

On April 26, 2021,23


In February 2022, we announced thatprovided the FDA acceptedwith an update on results from patients with CKD caused by Alport syndrome in the ongoing EAGLE trial. Mean increases in eGFR were observed at Week 12, Week 24, and Week 48 relative to Day 0 (before treatment) in EAGLE in patients who previously received placebo and initiated treatment with bardoxolone in EAGLE. Patients who previously received bardoxolone for filingtwo years in CARDINAL experienced similar mean increases in eGFR at all timepoints. For the 37 patients randomized to bardoxolone in CARDINAL who completed 48 weeks in EAGLE, bardoxolone treatment resulted in a mean on-treatment change from baseline in eGFR (relative to original CARDINAL baseline) of 9.2 mL/min/1.73 m2 at Year 1, 7.8 mL/min/1.73 m2 at Year 2, and 6.7 mL/min/1.73 m2 at Year 3.

A subset of patients with Alport syndrome (n=18) completed 96 weeks of treatment in EAGLE, which amounts to approximately four years of total treatment, and had a mean ± standard error change in eGFR from CARDINAL baseline of 5.5 ± 3.5 mL/min/1.73 m2. This sustained improvement of kidney function is notable when compared to the CARDINAL trial population’s expected yearly eGFR decline of 5.1 mL/min/1.73 m2, which was calculated based on five-year historical eGFR data collected before patients entered the trial. No new safety findings have been identified in the EAGLE trial.

On February 25, 2022, we received a CRL from the FDA with respect to its review of our NDA for bardoxolone forin the treatment of patients with CKD caused by Alport syndrome. The CRL indicated that the FDA willcannot approve the NDA in its present form. Based on its review, the application under a Standard Review timeline.  The PDUFA date, the FDA action date for the application, is scheduled for February 25, 2022.  The FDA also advised usconcluded that it does not believe the submitted data demonstrates that bardoxolone is currently planningeffective in slowing the loss of kidney function in patients with Alport syndrome and reducing the risk of progression to hold ankidney failure and has requested additional data to support the efficacy and safety of bardoxolone. Their conclusion was based on efficacy and safety concerns primarily set forth in the FDA’s briefing book and discussed at the Cardiovascular and Renal Drugs Advisory Committee meeting held on December 8, 2021.

The FDA stated that the issues could be resolved by providing evidence of effectiveness that includes evidence from an adequate and well-controlled study showing a clinically relevant effect on the rate of loss of kidney function in patients with Alport syndrome or, alternatively, an effect on a clinical outcome (i.e., an endpoint that captures how patients with Alport syndrome feel, function, or survive). In addition, the FDA stated that we would need to discussaddress whether bardoxolone has a clinically relevant effect on the application.  We also plan to pursue marketing approval outsideQT interval and show that the demonstrated clinical benefits of bardoxolone outweigh its risks. The FDA welcomed continued discussion on the United States.details of a path forward. We plan to submit a MAAwork closely with the EMAFDA to achieve our goal of bringing this important medicine to patients in the fourth quarter ofUnited States.

In October 2021, we submitted an MAA to the EMA for marketing approval of bardoxolone forin the treatment of patients with CKD caused by Alport syndromesyndrome. In the first quarter of 2022, we received the 120-day list of questions from the EMA. We are in the European Union.process of preparing our responses. We requested a 90-day extension for our responses which was granted by the EMA. The timeline for the EMA’s review cycle was therefore extended.


Bardoxolone in Patients with ADPKDAutosomal Dominant Polycystic Kidney Disease

ADPKD is a rare and serious hereditary form of CKD caused by a genetic defect in PKD1 or PKD2 genes leading to the formation of fluid-filled cysts in the kidneys and other organs. Cyst growth can cause the kidneys to expand up to five to seven times their normal volume, leading to pain and progressive loss of kidney function. Inflammation appears to play a role in cyst growth and is associated with disease progression in ADPKD.

ADPKD affects both men and women of all racial and ethnic groups and is the leading inheritable cause of kidney failure with an estimated diagnosed population of 140,000 patients and an estimated prevalent population of 400,000 patients in the United States. Despite current standard of carestandard-of-care treatment, an estimated 50% of ADPKD patients progress to ESKD and require dialysis or a kidney transplant by 60 years of age. The only therapy currently approved for ADPKD is JYNARQUE® (tolvaptan), developed by Otsuka Pharmaceuticals Co., Ltd., which was approved in the United States in 2018 to slow kidney function decline in adults at risk of rapidly progressing ADPKD.

We are currently enrolling patients in FALCON, an international, multi-center, randomized, double-blind, placebo-controlled trial studying the safety and efficacy of bardoxolone in patients with ADPKD randomized one-to-one to bardoxoloneactive drug or placebo. FALCON will enrollis enrolling patients in a broad range of ages, 18 to 70 years old, with an eGFR between 30 toand 90 mL/min/1.73 m2.

24


In the firstquarter of 2022, we finalized a protocol amendment to FALCON, and we initiated submission of the amendment to the FDA and other relevant health authorities. The major protocol amendment changes include an increase in the sample size from 550 to 850 patients, addition of adolescent (12 to 17 years) patients with ADPKD for a total age range of 12 to 70 years, removal of the off-treatment period (Week 48 – Week 52) during Year 1, change of the primary endpoint is theof off-treatment eGFR change from baseline versus placebo at Week 52 which represents 48(or four weeks after drug discontinuation in Year 1) to eGFR change from baseline at Week 108 (eight weeks after planned drug discontinuation at Week 100). The protocol amendment also includes addition of an exploratory endpoint of eGFR change from baseline at Week 112 (12 weeks after planned drug discontinuation at Week 100), and addition of a sub study with ambulatory blood pressure monitoring.

As agreed with the FDA prior to submission of the amendment, we recently had a Type A meeting to discuss key changes made to the FALCON protocol. Based on the discussion during the meeting and the meeting minutes, the Division stated that the proposed primary endpoint of eGFR change from baseline at Week 108 (8 weeks after planned drug discontinuation at Week 100) was reasonable since the available data suggest that bardoxolone’s acute pharmacodynamic effect on eGFR should be largely resolved. The Division provided guidance on handling of data from patients who completed Year 2 of the study before the protocol amendment and so did not have an eGFR assessment at Week 108. This included direction on imputing missing data for these patients in the primary analysis. The Division stated that, in addition to the primary endpoint, it will be important to demonstrate that the treatment effect accrues over time to support a claim that bardoxolone slows the loss of kidney function in patients with ADPKD and provided guidance on the statistical methodologies for the exploratory eGFR slope analyses. The FDA also confirmed that if FALCON is positive, it could support registration of bardoxolone in ADPKD.

Pursuant to the protocol amendment, patients will be treated with bardoxolone or placebo for 100 weeks followed by a four-weektwelve-week withdrawal period. AtThe trial will remain blinded until study completion. All patients will be asked to return at Week 52, patients resume treatment for a second 48-week period to Week 100 followed by a second four-week withdrawal period to Week 104.108 independent of the time of study drug discontinuation. The key secondary endpoint is the off-treatment eGFR change from baseline versus placebo at Week 104.

In the first quarter of 2021, we amended the FALCON protocol to increase the target enrollment from 300 patients to a total of approximately100. More than 550 patients. We observed an increase in variability in eGFR values in Year 2 of the CARDINAL study versus Year 1. The increase in enrollment is intended to preserve the statistical power of the study in the event that we observe similar eGFR variability to what we observed in Year 2 of the CARDINAL study. The FDA has provided us with guidance that, in patients with ADPKD, an analysis of eGFR during the off-treatment period demonstrating an improvement versus placebo after one year of bardoxolone treatment may support an NDA submission for accelerated approval of bardoxolone for the treatment of ADPKD, and data demonstrating an improvement versus placebo after two years of treatment may support full approval.

In March 2020, we announced a temporary pause in enrollment in FALCON due to the COVID-19 pandemic; we resumed enrollment during the third quarter of 2020. The measures we implemented to the conduct of FALCON in response to COVID-19 have been effective, and we anticipate no meaningful impact on data integrity due to COVID-19. Most sites are currently able to screen and randomize patients, and more than 290 patients are currently enrolled in the study. We expect to complete enrollmenttrial.

AYAME Trial in the FALCON studyDiabetic CKD Conducted by the endKyowa Kirin

Upon completion of 2021.

Bardoxolone in Patients with CKD at Risk of Rapid Progression

MERLIN is a proof of concept, multi-center, double-blind, placebo-controlled,Kyowa Kirin's Phase 2 TSUBAKI trial to evaluate the safety and efficacy of bardoxolone in patient populations with CKD at meaningful risk of progression to ESKD. Multiple etiologies of CKD will be studied, including patient populations we have studied before (Alport syndrome, IgAN, FSGS, T1D CKD, and T2D CKD) and those we have not studied, such as hypertensive CKD and others.

MERLIN is anticipated to enroll approximately 70 patients with eGFR between 20Stage 3 and 60 mL/min/1.73 m2,4 diabetic CKD in Japan, and after discussions with the Pharmaceuticals and Medical Devices Agency, Kyowa Kirin initiated a Phase 3 outcomes trial called AYAME in patients must meet at least one of the following criteria: UACR ≥ 300 mg/g; eGFR decline at a rate of ≥with Stage 3 or 4 mL/min/1.73 m2diabetic CKD in prior year; or hematuria defined as > 5-10 red blood cells per high power field (manual method), documented history of positive urinary dipstick for blood in prior year, or macroscopic hematuria in prior 3 years. We expect to complete enrollment in the MERLIN trial by the end of the second quarter of 2021, and we expect data in the second half of 2021.

Japan. The primary objectiveendpoint is time to assessonset of a ≥ 30% decrease in eGFR from baseline or ESKD. The secondary endpoints are time to onset of a ≥ 40% decrease in eGFR from baseline or ESKD, time to onset of a ≥ 53% decrease in eGFR from baseline or ESKD, time to onset of ESKD, and change in eGFR from baseline at week 12,each evaluation time point. Kyowa Kirin completed patient enrollment in AYAME in June 2019 and expects the secondary objective islast patient visit to characterize change in eGFR from baseline by CKD etiology at Week 12. The exploratory efficacy objective is to characterize change in eGFR from baseline during a 5-week drug treatment withdrawal period. The results from MERLIN would provide us proof of concept to potentially proceed to a larger Phase 3 study with similar eligibility criteria and a longer treatment duration. Moreover, together with the results of KKC’s ongoing AYAME study, which are expectedoccur in the firstsecond half of 2022, MERLIN will inform us2022.

U.S. Commercial Readiness

With the NDA submission of omaveloxolone complete, we are advancing commercial launch preparations in the potential to commercially pursue bardoxoloneUnited States. We are in broad setthe process of CKD patients at risk for rapid progression.


Both the FALCON and MERLIN trials draw from the results of our Phase 2 study called PHOENIX, an open-label, multi-center Phase 2 trial evaluating the safety and efficacy of bardoxolone in patients with ADPKD, IgAN, T1D CKD, or FSGS completed in 2019. In each of these cohorts, patients treated with bardoxolone experienced a statistically significant increase from baseline in mean eGFR after 12 weeks of treatment. The most commonly reported AE across all cohorts was muscle spasms, which were not associated with clinical signs or symptoms of muscle injury. The overall rate of SAEs was low, with three patients reporting treatment-emergent SAEs, none of which were reported as related to bardoxolone.  We plan to pursue development opportunities in each of these rare and serious forms of CKD, maintaining our intent to expandbuilding the commercial indications for bardoxolone.

Historical Development of Bardoxolone

Priorinfrastructure necessary to our CARDINAL Phase 3 trial, clinical trials enrolling over 2,000 patients exposed to bardoxolone have demonstrated consistent, clinically meaningful improvement in kidney function across several disease states as measured by eGFR and other markers of kidney function. Specifically, we have observed statistically significant increases in eGFR in all Phase 2 and Phase 3 clinical trials in seven distinct patient populations treated with bardoxolone, including patients with pulmonary hypertension and CKD caused by T2D CKD, Alport syndrome, ADPKD, IgAN, T1D CKD, and FSGS.

We believe these data, in addition to the CARDINAL Phase 3 data,effectively support the potential for bardoxolone to delay or prevent GFR declines that cause the need for dialysis or kidney transplant, and eventually death in patients with Alport syndrome and other formscommercialization of CKD.

Additional observations from the prior clinical trials of bardoxolone include the following:

Statistically significant increases in directly-measured GFR using the “gold standard” inulin clearance method, improvements in creatinine clearance, and reduction in the levels of blood waste products filtered by the kidney.

Statistically significant improvements in eGFR versus baseline or placebo in six different types of CKD, including Alport syndrome, ADPKD, IgAN, T1D CKD, T2D CKD, and FSGS.

Sustained improvement in kidney function in long-term trials:

o

In the Phase 2 portion of CARDINAL, bardoxolone treatment produced a statistically significant increase from baseline in mean eGFR of 10.4 mL/min/1.73 m2 (p<0.0001) after 48 weeks of treatment, which, based on historical data available for 22 of the patients prior to enrolling in the trial, represents a recovery of over two years of average decline in kidney function.

o

In two large, placebo-controlled clinical studies (BEAM and BEACON) in patients with T2D CKD, statistically significant increases in mean eGFR of 14.9 mL/min/1.73 m2 (p<0.001) and 5.6 mL/min/1.73 m2 (p<0.001), respectively, were sustained for at least one year.

Reduction in risk of adverse kidney outcomes, suggesting that bardoxolone treatment preserves kidney function and may delay the onset of kidney failure in patients with T2D and stage 4 CKD:

o

In BEACON, patients randomized to bardoxolone were significantly less likely to experience adverse kidney outcomes as defined by a composite endpoint consisting of ≥30% decline from baseline in eGFR, eGFR <15 mL/min/1.73 m2, or ESKD events (HR=0.48, p<0.0001).  

o

In BEACON, bardoxolone treatment resulted in a decreased number of kidney-related SAEs and ESKD events.  

Statistically significant improvement in eGFR above baseline or versus placebo during the off-treatment period in BEAM, BEACON, the Phase 2 portion of CARDINAL at one year, and the Phase 3 portion of CARDINAL at one and two years.

o

The FDA has provided guidance to us and other sponsors that clinical trials with an eGFR benefit versus placebo during the off-treatment period may support approval in certain rare forms of CKD.

o

We believe that the increase in off-treatment eGFR relative to placebo shows that increases in eGFR due to bardoxolone over longer durations do not have detrimental effects on kidney function. Most importantly, the observed eGFR benefit versus placebo during the off-treatment periods in these clinical trials demonstrates that bardoxolone treatment may have resulted in


structural improvement, modifying the course of the disease, and delaying the need for dialysis or kidney transplant.

Programs in Neurological Diseases

Omaveloxolone in Patients with FA

We are developing omaveloxolone for the treatment of patientsFA, if and when we receive regulatory approval. Commercial launch preparation for omaveloxolone in FA will continue to advance in step with FA,the regulatory progress.

Our ability to launch omaveloxolone is dependent on the successful filing and defense of an inherited, debilitating,NDA and degenerative neuromuscular disorderapproval by the FDA. We have hired commercial leadership and intend to build the teams, infrastructure, systems, and processes necessary for the launch of omaveloxolone. This will include sales, marketing, market access, patient support, and distribution. Additionally, we are expanding quality and compliance functions to support commercialization. A trade name for omaveloxolone has been selected.

25


One challenge unique to rare-disease commercialization is patient identification due to the very small and sometimes heterogeneous disease populations. Our management team is experienced in maximizing patient identification for both clinical development and commercialization purposes in rare diseases. Commercial infrastructure for orphan products typically consists of a targeted, specialty sales force that calls on a limited and focused group of physicians and personnel involved in sales management, internal sales support, marketing, patient access, and distribution.

Ex-U.S. Commercial Readiness

Our ability to launch omaveloxolone is normally diagnosed during adolescencedependent on the successful filing and can lead to premature death.  Patients with FA experience progressive lossdefense of coordination, muscle weakness,an MAA and fatigue, which commonly progresses to motor incapacitation and wheelchair reliance.  Symptoms generally occur in children, with patients requiring a wheelchairapproval by their teensEMA or early twenties.  FA affects approximately 5,000 children and adults inother regulatory agencies. Outside of the United States, where appropriate and 22,000 individuals globally.  Theredepending on the terms of our contractual arrangements, we plan, either alone, or with new collaboration partners, to commercialize our products. Our strategic collaborator Kyowa Kirin has all rights to commercialize bardoxolone in its territories. We are currently no approved therapiesrefining our strategy and market assessments with respect to treat FA.

Our Phase 2 trial, called MOXIe, was a two-part, international, multi-center, randomized, double-blind, placebo-controlled registrational trial that studiedpotential launches in the safetyEU, and efficacy ofwe continue to evaluate market opportunities for our products in other global markets. Commercial launch preparation for omaveloxolone in patients with FA.  Additionally, patients who completed the study and met eligibility requirements could participate in the MOXIe Extension during which investigators and patients remained blinded to prior treatment assignments.  In October 2019, we announced that Part 2 in patients with FA met its primary endpointoutside of change in mFARS relative to placebo after 48 weeks of treatment.  Patients treated with omaveloxolone (150 mg/day) demonstrated a statistically significant, placebo-corrected 2.40 point mean improvement (decrease) in mFARS after 48 weeks of treatment (p=0.014).  Patients treated with omaveloxolone demonstrated improvement relative to placebo in every subcategory measured under mFARS.  Omaveloxolone treatment was generally reported to be well-tolerated.  

At a Type C meeting in August 2020, the FDA provided us guidance that, although it does not have concerns with the reliability of the mFARS primary endpoint results from MOXIe Part 2, it was not convinced that the results from MOXIe Part 2, as a single study, were sufficient to support approval. The FDA stated that we will need to conduct a second pivotal trial that confirms the mFARS results of MOXIe Part 2 with a similar magnitude of effect. As an alternative, we proposed the Baseline-Controlled Study.

The Baseline-Controlled Study evaluated the efficacy of omaveloxolone treatment using a baseline-controlled analysis design in which patients serve as their own controls, and the annualized rate of change in mFARS during the pre-treatment period in either MOXIe Part 1 or Part 2 was compared to annualized rate of change in mFARS during the treatment period in the MOXIe Extension for patients who received approximately 48 weeks of omaveloxolone in the MOXIe Extension (the paired difference).  The Baseline-Controlled Study demonstrated a statistically significant -3.76 point improvement (p=0.0022) for the primary endpoint of the paired difference in annualized mFARS slopes between the treatment and pre-treatment periods in the primary analysis population.

These results were provided to the FDA, and after an internal review, the FDA concluded that it does not believe the results strengthen the results of MOXIe Part 2.  The FDA suggested additional exploratory analyses to evaluate whether the treatment effects show an effect on disease course using patients randomized to placebo during MOXIe Part 2 who then went on study drug in the MOXIe Extension. The FDA noted the small number of patients and stated that the potential for these exploratory analyses to strengthen the study results was questionable.  The FDA indicated that it remains interested in reviewing the results of the additional exploratory analyses, called the Delayed-Start Analyses, as those may inform the future development program.  

The Delayed-Start Analyses included the change from baseline in mFARS during the MOXIe Extension, comparing patients randomized to placebo (the placebo-to-omaveloxolone group) with those randomized to omaveloxolone (the omaveloxolone-to-omaveloxolone group) in the double-blind period from MOXIe Part 2. Such analyses would include a graphical representation of the time course for the change from baseline mFARS in both omaveloxolone and placebo groups from the placebo-controlled MOXIe Part 2 and the change from baseline in the two treatments groups (the omaveloxolone-to-omaveloxolone group and the placebo-to-omaveloxolone group) through 48 weeks in the MOXIe Extension.


A total of 73 out of 75 (97%) patients without pes cavus who completed MOXIe Part 2 enrolled in the MOXIe Extension, including 39 patients previously randomized to placebo (the placebo-to-omaveloxolone group) and 34 patients previously randomized to omaveloxolone (the omaveloxolone-to-omaveloxolone group). A longitudinal analysis used to calculate annualized slopes incorporating all available data from the MOXIe Extension showed similar slopes in mFARS for the placebo-to-omaveloxolone group (0.59 points per year) when compared to the omaveloxolone-to-omaveloxolone group (0.41 points per year) with no significant difference between slopes (p=0.75). The resulting parallel trajectories between both treatment groups is consistent with disease-modifying activity.

We believe that the results of the Delayed-Start Analyses suggest disease-modifying activity with omaveloxolone, provide evidence supporting the positive primary endpoint findings in MOXIe Part 2, and provide additional evidence of the effectiveness of omaveloxolone in FA. We requested and the FDA has granted us a Type C meeting, which is scheduled to occur in the second quarter of 2021, to discuss the Delayed-Start Analyses and the FA development program. We plan to initiate a second pivotal study in the fourth quarter of 2021, incorporating input from both the FDA and the EMA into the protocol before we initiate enrollment.

Omaveloxolone in Other Potential Indications

Omaveloxolone is a promising platform molecule. Because mitochondrial dysfunction is a key feature of many neurological and neuromuscular diseases, we believe omaveloxolone may be broadly applicable to treat such diseases by activating Nrf2 to normalize and improve mitochondrial function and ATP production.

Based on our understanding of the pathophysiology of neurological diseases, characterized by mitochondrial dysfunction, inflammation, and oxidative stress, we believe omaveloxolone may be applicable to diseases such as progressive supranuclear palsy, Parkinson’s disease, frontotemporal dementia, Huntington’s disease, ALS, Alzheimer’s disease, and epilepsy. Consistent with this, we have observed compelling activity of omaveloxolone and our other Nrf2 activators in preclinical models of many of these diseases.

Our Nrf2 activators reduced seizure frequency in refractory, progressive epilepsy models and restored mitochondrial function in patient biopsy samples and preclinical models of FA, ALS, familial and sporadic Parkinson’s disease, and frontotemporal dementia. In clinical trials, improvements in neuromuscular function have been observed in FA patients treated with omaveloxolone as assessed by mFARS rating scale, and improvements in mitochondrial function, as measured by reductions in blood lactate and heart rate, have been observed in patients with primary mitochondrial disease. Accordingly, we believe that omaveloxolone has the potential to treat a number of neurological and neuromuscular diseases that currently have few or no effective therapies, and we plan to pursue the development of omaveloxolone and our other Nrf2 activators for one or more of these diseases.

RTA 901 in Neurodegeneration and Neuroprotection Diseases

RTA 901 is the lead product candidate from our Hsp90 modulator program, which includes highly potent and selective C-terminal modulators of Hsp90. We have observed favorable activity of RTA 901 in a range of preclinical models of neurological disease, including models of diabetic neuropathy, neuroinflammation, and neuropathic pain.

Historically, other companies have explored N-terminal Hsp90 inhibitors for cancer therapeutics; however, this approach has been associated with multiple adverse effects including peripheral neuropathy and ocular toxicity. Binding at the C-terminus of Hsp90 leads to increased transcription of Hsp70, a cytoprotective and molecular chaperone gene, which facilitates cell survival in response to stress without the deleterious activities of N-terminal inhibition.

In preclinical rodent disease models, we observed that RTA 901 administered orally once-daily rescued existing nerve function, restored thermal and mechanical sensitivity within four weeks, and improved nerve conductance velocity and mitochondrial function. These effects are dose-dependent, reversible, and HSP70-dependent.


We completed a Phase 1 single and multiple ascending dose trial of oral, once-daily RTA 901 in healthy adult volunteers to evaluate the safety, tolerability, and pharmacokinetic (PK) profile. The PK was linear up to the highest doses evaluated with a half-life ranging from two to nine hours, and exposures were easily achievable in 10-fold excess of those necessary for efficacy in multiple animal models. No safety or tolerability concerns were reported. We plan to initiate additional Phase 1 studies to evaluate the PK and drug-drug interaction potential of RTA 901 in the second quarter of 2021, and we expect to launch a randomized, placebo-controlled Phase 2 study in diabetic peripheral neuropathic pain (DPNP) in the fourth quarter of 2021.

There are about four million patients with moderate to severe DPNP in the United States and about two million adult patients diagnosedwill advance in step with DPNP seek treatment annually. We are the exclusive licensee of RTA 901 and maintain worldwide commercial rights.regulatory progress.

Other Clinical Programs

In addition to our lead programs, we are developing RTA 1701, the lead product candidate from our proprietary series of RORγt inhibitors, for the potential treatment of a broad range of autoimmune, inflammatory, and fibrotic diseases.   RTA 1701 is an orally-bioavailable, RORγt-selective allosteric inhibitor that suppresses Th17 differentiation in vitro and demonstrates strong efficacy in rodent disease models of autoimmune disease. RTA 1701 also potently suppresses the production of IL-17A, a clinically important cytokine, in human immune cells and when dosed orally to non-human primates. We have conducted a Phase 1 trial to evaluate the safety, tolerability, and PK profile of RTA 1701 in healthy adult volunteers. No safety or tolerability concerns were reported, and we observed an acceptable PK profile. We plan to continue development of RTA 1701 in autoimmune, inflammatory, or fibrotic diseases. We retain all rights to our RORγt inhibitors, which are not subject to any existing commercial collaborations.


Corporate Overview

To date, we have focused most of our efforts and resources on developing our product candidates and conducting preclinical studies and clinical trials. We have historically financed our operations primarily through revenue generated from our collaborations with AbbVie and KKC,Kyowa Kirin, from sales of our securities, secured loans, and a strategic financing from BXLS. We have not received any payments or revenue from collaborations other than nonrefundable upfront, milestone, and cost sharing payments from our collaborations with AbbVie and KKC,Kyowa Kirin, from the Development Agreement with BXLS, and from reimbursements of expenses under the terms of our agreement with KKC.Kyowa Kirin. We have incurred losses in each year since our inception, other than in 2014. As of March 31, 2021,2022, we had $777.6$532.0 million of cash and cash equivalents and an accumulated deficit of $1,025.7$1,329.5 million. We continue to incur significant research and development and other expenses related to our ongoing operations. Despite contractual product development commitments and the potential to receive future payments from KKC,Kyowa Kirin, we anticipate that we will continue to incur losses for the foreseeable future, and we anticipate that our losses will increase as we continue our development of, seek regulatory approval for, and potential commercialization ofpotentially commercialize our product candidates. If we do not successfully develop and obtain regulatory approval of our existing product candidates or any future product candidates and effectively manufacture, market, and sell any products that are approved, we may never generate revenue from product sales. Furthermore, even if we do generate revenue from product sales, we may never again achieve or sustain profitability on a quarterly or annual basis. Our prior losses, combined with expected future losses, have had and will continue to have an adverse effect on our stockholders’ equity and working capital. Our failure to become and remain profitable could depress the market price of our Class A common stock and could impair our ability to raise capital, expand our business, diversify our product offerings, or continue our operations.

The probability of success for each of our product candidates and clinical programs and our ability to generate product revenue and become profitable depend upon a variety of factors, including the quality of the product candidate, clinical results, investment in the program, competition, manufacturing capability, commercial viability, and our collaborators’ ability to successfully execute our development and commercialization plans. We will also require additional capital through equity, debt, or royalty financings or collaboration arrangements in order to fund our operations and execute on our business plans, and there is no assurance that such financing or arrangements will be available to us on commercially reasonable terms or at all. For a description of the numerous risks and uncertainties associated with product development and raising additional capital, see “Risk Factors” included in our Annual Report on Form 10-K for the year ended December 31, 2021.

26


Financial Operations Overview

Revenue

Our revenue to date has been generated primarily from licensing fees received under our collaborative license agreements and reimbursements for expenses. We currently have no approved products and have not generated any revenue from the sale of products to date. In the future, we may generate revenue from product sales, royalties on product sales, reimbursements for collaboration services under our current collaboration agreements, or license fees, milestones, or upfront payments if we enter into any new collaborations or license agreements. We expect that our future revenue will fluctuate from quarter to quarter for many reasons, including the uncertain timing and amount of any such payments and sales.

Our license and milestone revenue has been generated primarily from the KKCKyowa Kirin Agreement, the AbbVie License Agreement, and the Collaboration Agreement and consists of upfront payments and milestone payments. License revenue recorded with respect to the KKCKyowa Kirin Agreement, the AbbVie License Agreement, and the AbbVie Collaboration Agreement consists solely of the recognition of deferred revenue. Under our revenue recognition policy, collaboration revenue associated with upfront, non-refundable license payments received under our license and collaboration agreements are deferred and recognized ratably over the expected term of the performance obligations under each agreement. Under the Reacquisition Agreement, we no longer have performance obligations under the AbbVie License Agreement and the Collaboration Agreement. WeUnder the Kyowa Kirin Agreement, we only expect to recognize the deferred revenue under the KKC Agreement through mid-2022.June 2022.

Research and Development Expenses

The largest component of our total operating expenses has historically been our investment in research and development activities, including the clinical development of our product candidates. From our inception through March 31, 2021,2022, we have incurred a total of $968.8$1,129.7 million in research and development expense, a majority of which relates to the development of bardoxolone and omaveloxolone. We expect our research and development expense to continue to increase in the future as we advance our product candidates through clinical trials and expand our product candidate portfolio. The process of conducting the necessary clinical research to obtain


regulatory approval is costly and time-consuming, and we consider the active management and development of our clinical pipeline to be crucial to our long-term success. The actual probability of success for each product candidate and preclinical program may be affected by a variety of factors, including the safety and efficacy data for product candidates, investment in the program, competition, manufacturing capability, and commercial viability.

Research and development expenses include:

expenses incurred under agreements with clinical trial sites that conduct research and development activities on our behalf;
expenses incurred under contract research agreements and other agreements with third parties;
employee and consultant-related expenses, which include salaries, benefits, travel, and stock-based compensation;
laboratory and vendor expenses related to the execution of preclinical and non-clinical studies and clinical trials;
the cost of acquiring, developing, manufacturing, and distributing clinical trial materials;
the cost of development, scale up, and process validation activities to support product registration; and
facilities, depreciation, and other expenses, which include direct and allocated expenses for rent and maintenance of facilities, insurance, and other supply costs.

expenses incurred under agreements with clinical trial sites that conduct research and development activities on our behalf;

expenses incurred under contract research agreements and other agreements with third parties;

employee and consultant-related expenses, which include salaries, benefits, travel, and stock-based compensation;

laboratory and vendor expenses related to the execution of preclinical and non-clinical studies and clinical trials;

the cost of acquiring, developing, manufacturing, and distributing clinical trial materials;

the cost of development, scale up, and process validation activities to support product registration; and

facilities, depreciation, and other expenses, which include direct and allocated expenses for rent and maintenance of facilities, insurance, and other supply costs.

Research and development costs are expensed as incurred. Costs for certain development activities such as clinical trials are highly judgmental and are recognized based on an evaluation of the progress to completion of specific tasks using information and data provided to us by our vendors and our clinical sites.

27


We base our expense accruals related to clinical trials on our estimates of the services received and efforts expended pursuant to contracts with multiple research institutions and CROs that conduct and manage clinical trials on our behalf. The financial terms of these agreements vary from contract to contract and may result in uneven payment flows. Payments under some of these contracts depend on factors such as the successful enrollment of patients and the completion of clinical trial milestones. In accruing costs, we estimate the time period over which services will be performed and the level of effort to be expended in each period.  If we do not identify costs that we have begun to incur or if we underestimate or overestimate the level of services performed or the costs of these services, our actual expenses could differ from our estimates.

To date, we have not experienced material changes in our estimates of accrued research and development expenses after a reporting period. However, due to the nature of estimates, we cannot assure you that we will not make changes to our estimates in the future as we become aware of additional information about the status or conduct of our clinical trials and other research activities.

Currently, KKCKyowa Kirin has allowed us to conduct clinical studies of bardoxolone in certain rare forms of kidney diseases in Japan and has reimbursed us the majority of the costs for our CARDINAL study in Japan. Kyowa Kirin is the in-country caretaker in our FALCON study in Japan and is payingwe are reimbursing Kyowa Kirin for the costs of a certain number of patients asin the in-country caretaker in our FALCON study in Japan.study.

The following table summarizes our research and development expenses incurred:incurred during the three months ended March 31:

 

Three Months Ended

 

 

March 31

 

 

2021

 

 

2020

 

 

2022

 

 

2021

 

 

(in thousands)

 

 

(in thousands)

 

Bardoxolone

 

$

11,386

 

 

$

13,962

 

 

$

6,462

 

$

11,386

 

Omaveloxolone

 

 

2,473

 

 

 

6,674

 

 

7,597

 

2,473

 

RTA 901

 

 

706

 

 

 

1,211

 

 

1,407

 

706

 

RTA 1701

 

 

177

 

 

 

1,395

 

Other research and development expenses

 

 

20,138

 

 

 

24,411

 

Other research and development expenses (1)

 

 

24,338

 

 

 

20,315

 

Total research and development expenses

 

$

34,880

 

 

$

47,653

 

 

$

39,804

 

 

$

34,880

 

(1) RTA 1701 expenses have been included in other research and development expenses due to development updates in the program.

The program-specific expenses summarized in the table above include costs that we directly allocate to our product candidates. Our other research and development expenses include research and development salaries, benefits, stock-based compensation and preclinical, research, and discovery costs, which we do not allocate on a program-specific basis.

General and Administrative Expenses

General and administrative expenses consist primarily of employee-related expenses for executive, operational, finance, legal, compliance, and human resource functions. Other general and administrative expenses include personnel expense, facility-related costs, professional fees, accounting and legal services, depreciation expense, other external services, and expenses associated with obtaining and maintaining our intellectual property rights.

We anticipate that our general and administrative expenses will increase in the future as we prepare forincrease our headcount to support our continued research and development and potential commercialization of our product candidates. We have also incurred, and anticipate incurring in the future, increased expenses associated with being a public company, including exchange listing and SEC requirements, director and officer insurance premiums, legal, audit and tax fees, compliance with the Sarbanes-Oxley Act, of 2002, regulatory compliance programs, and investor relations costs. Additionally, if and when we believe the first regulatory approval of one of our product candidates appears likely, we anticipate an increase in payroll and related expenses as a result of our preparation for commercial operations, especially for the sales and marketing of our product candidates.

Other Income (Expense), Net

Other income (expense) includes interest and gains earned on our cash and cash equivalents, interest expense on our Term Loans, amortization of debt issuance costs, imputed interest on long term payables, loss on extinguishment of debt, gain on termination of lease, foreign currency exchange gains and losses, gains and losses on sales of assets, and non-cash interest expense on liability related to the sale of future royalties.

28


Benefit from (Provision for) Taxes on Income

Provision for taxes on income consists of net loss, taxed at federal tax rates and adjusted for certain permanent differences. Realization of deferred tax assets is generally dependent upon future earnings by jurisdiction, of which the timing and amount are uncertain for the majority of our deferred tax assets, and valuation allowances are maintained against them. Changes in valuation allowances also affect the tax provision.


Results of Operations

Comparison of the Three Months Ended March 31, 2022 and 2021 and 2020 (unaudited)

The following table sets forth our results of operations for the three months ended March 31:

 

2022

 

 

2021

 

 

Change $

 

 

Change %

 

 

2021

 

 

2020

 

 

Change $

 

 

Change %

 

 

(in thousands, except for percentage data)

 

Collaboration revenue

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

License and milestone

 

$

795

 

 

$

1,169

 

 

$

(374

)

 

 

(32

)

 

$

893

 

$

795

 

$

98

 

12

 

Other revenue

 

 

149

 

 

 

184

 

 

 

(35

)

 

 

(19

)

 

 

21

 

 

 

149

 

 

 

(128

)

 

 

(86

)

Total collaboration revenue

 

 

944

 

 

 

1,353.0

 

 

 

(409

)

 

 

(30

)

 

914

 

944

 

(30

)

 

(3

)

Expenses

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Research and development

 

 

34,880

 

 

 

47,653

 

 

 

(12,773

)

 

 

(27

)

 

39,804

 

34,880

 

4,924

 

14

 

General and administrative

 

 

20,704

 

 

 

20,787

 

 

 

(83

)

 

 

(0

)

 

24,841

 

20,704

 

4,137

 

20

 

Depreciation

 

 

274

 

 

 

278

 

 

 

(4

)

 

 

(1

)

 

 

308

 

 

 

274

 

 

 

34

 

 

 

12

 

Total expenses

 

 

55,858

 

 

 

68,718

 

 

 

(12,860

)

 

 

(19

)

 

64,953

 

55,858

 

9,095

 

16

 

Other income (expense), net

 

 

(12,556

)

 

 

(3,814

)

 

 

(8,742

)

 

**

 

 

 

(9,772

)

 

 

(12,556

)

 

 

2,784

 

 

 

22

 

Loss before taxes on income

 

 

(67,470

)

 

 

(71,179

)

 

 

3,709

 

 

 

5

 

 

(73,811

)

 

(67,470

)

 

(6,341

)

 

(9

)

Benefit from (provision for) taxes on income

 

 

15

 

 

 

22,240

 

 

 

(22,225

)

 

 

(100

)

 

 

(31

)

 

 

15

 

 

 

(46

)

 

**

 

Net loss

 

$

(67,455

)

 

$

(48,939

)

 

$

(18,516

)

 

 

(38

)

 

$

(73,842

)

 

$

(67,455

)

 

$

(6,387

)

 

 

(9

)

** Percentage not meaningful

Revenue

License and milestone revenue represented approximately 84%98% and 86%84% of total revenue for the three months ended March 31, 20212022 and 2020,2021, respectively, and consisted of the recognition of KKCKyowa Kirin deferred revenue. License and milestone revenue decreasedincreased by $0.4 million or 32%12% during the three months ended March 31, 2021,2022, compared to the three months ended March 31, 2020, 2021, primarily due to the extensionachievement of our performance obligation perioda regulatory milestone in July 2021, variable consideration previously considered constrained, under the KKCKyowa Kirin Agreement from December 2021 to June 2022 during the three months ended March 31, 2021. Accordingly, the quarterly recognition of the deferred revenue under the KKC Agreement was reduced from $1.2 million to $0.8 million..

Other revenue was immaterial for the three months ended March 31, 20212022, and 2020.2021.

Expenses

The following table summarizes our expenses, including as a percentage of total expenses, for the three months ended March 31:

 

2021

 

 

% of Total

Expenses

 

 

2020

 

 

% of Total

Expenses

 

 

2022

 

 

% of Total
Expenses

 

 

2021

 

 

% of Total
Expenses

 

 

(in thousands)

 

 

(in thousands, except for percentage data)

 

Research and development

 

$

34,880

 

 

 

62

%

 

$

47,653

 

 

 

69

%

 

$

39,804

 

61

%

 

$

34,880

 

62

%

General and administrative

 

 

20,704

 

 

 

37

%

 

 

20,787

 

 

 

30

%

 

24,841

 

38

%

 

20,704

 

37

%

Depreciation

 

 

274

 

 

 

1

%

 

 

278

 

 

 

1

%

 

 

308

 

 

1

%

 

 

274

 

 

1

%

Total expenses

 

$

55,858

 

 

 

 

 

 

$

68,718

 

 

 

 

 

 

$

64,953

 

 

 

 

 

$

55,858

 

 

 

 

29


Research and Development Expenses

Research and development expenses decreasedincreased by $12.8$4.9 million, or 27%14%, for the three months ended March 31, 2021,2022, compared to the three months ended March 31, 2020.2021. The decreaseincrease was primarily due to decreased spend related to clinical study expenses related to our studies related to bardoxolone in pulmonary arterial hypertension (PAH) that were terminated in the first quarter of 2020, manufacturing expenses related to omaveloxolone, and completion of RTA 1701 toxicology studies offset by the increase in regulatory costs to support the submission of our NDA. The remaining changes included decreased equity compensation expense due to accelerated expense recognized during the first quarter of 2020, offset by an increase inincreased personnel and personnel-related costs to support the growth of ourproduct development activities.

Research and development expenses, as a percentage of total expenses, was 62%61% and 69%62% for the three months ended March 31, 2022 and 2021, respectively. The decrease of 1% was due to the proportionately larger increase in general and 2020, respectively.  administrative expenses, compared to research and development expenses.

General and Administrative Expenses

General and administrative expenses decreasedincreased by $0.1$4.1 million, or 20%, for the three months ended March 31, 2021,2022, compared to the three months ended March 31, 2020.  2021. The increase was primarily due to rent expense related to the new headquarters building lease that commenced in December 2021.

General and administrative expenses, as a percentage of total expenses, was 37%38% and 30%37%, for the three months ended March 31, 20212022 and 2020,2021, respectively. The increase of 1% was primarily due to the relative decreaseproportionately larger increase in general and administrative expenses, compared to research and development expenses.

Other Income (Expense), Net

Other income (expense), net increaseddecreased by $8.7$2.8 million for the three months ended March 31, 2021,2022, compared to the three months ended March 31, 2020.2021. The increasedecrease was primarily due to a decrease in effective interest rate in non-cash interest expense on liability related to the sale of future royalties.

We periodically reassess the expected royalty payments under the Development Agreement, and to the extent such payment is greater or less than the initial estimate, we adjust the amortization. Based on our review in the current first quarter of 2022, we lowered our previous estimate of future sales for which royalties will be paid. Accordingly, we have prospectively adjusted and decreased interest income earned due torecognized lower market rates, offset by decreasednon-cash interest expense on our Term Loans that were paid off in mid-2020.during the quarter ended March 31, 2022.

Benefit from (Provision for) Taxes on Income

Benefit from (Provision for) taxes on income decreased by $22.2 millionwas immaterial for the three months ended March 31, 2021, compared to the three months ended March 31, 2020, primarily due to a tax benefit recognized during the three months ended March 31, 2020.2022 and 2021.


30


Liquidity and Capital Resources

Since our inception, we have funded our operations primarily through collaboration and license agreements, the sale of preferred and common stock, secured loans, and the sale of future royalties.royalty interests, and secured loans. Through March 31, 2021,2022, we have raised gross cash proceeds of $476.6 million through the sale of convertible preferred stock and $780.0$785.0 million from payments under license and collaboration agreements. We also obtained $1,222.1 million in net proceeds from our initial public offering, follow-on offerings, and the sale of our Class A common stock under the Purchase Agreement, and $299.0 million in net proceeds from the sale of future royalties under the Development Agreement. We have not generated any revenue from the sale of any products. As of March 31, 2021,2022, we had available cash and cash equivalents of approximately $777.6$532.0 million. Our cash and cash equivalents are invested in accordance with our investment policy, primarily with a view to liquidity and capital preservation.

Cash Flows

The following table sets forth the primary sources and uses of cash for each of the three months ended March 31:

 

 

2021

 

 

2020

 

 

 

(in thousands)

 

 

 

 

 

Net cash (used in) provided by:

 

 

 

 

 

 

 

 

Operating activities

 

$

(45,011

)

 

$

(41,173

)

Investing activities

 

 

(193

)

 

 

(85

)

Financing activities

 

 

4,678

 

 

 

1,422

 

Net change in cash and cash equivalents

 

$

(40,526

)

 

$

(39,836

)

 

 

2022

 

 

2021

 

 

 

(in thousands)

 

 

 

 

 

Net cash (used in) provided by:

 

 

 

 

 

 

Operating activities

 

$

(58,185

)

 

$

(45,011

)

Investing activities

 

 

(288

)

 

 

(193

)

Financing activities

 

 

194

 

 

 

4,678

 

Net change in cash and cash equivalents

 

$

(58,279

)

 

$

(40,526

)

 

Operating Activities

Net cash used in operating activities was $58.2 million for the three months ended March 31, 2022, consisting primarily of a net loss of $73.8 million adjusted for non-cash items including stock-based compensation expense of $15.4 million, non-cash interest expense on liability related to sale of future royalty of $9.9 million, and a net decrease in operating assets and liabilities of $10.0 million. The significant items in the change in operating assets that impacted our use of cash in operations include a decrease of $4.5 million in accounts payable due to timing of payments, and a decrease of $8.9 million in direct research and other current and long-term liabilities, primarily due to annual bonus payments.

Net cash used in operating activities was $45.0 million for the three months ended March 31, 2021, consisting primarily of a net loss of $67.5 million adjusted for non-cash items including stock-based compensation expense of $14.7 million, non-cash interest expense on liability related to the sale of future royaltyroyalties of $10.9 million, depreciation, amortization of issuance costs, and imputed interest expense of $2.0 million, and a net increase in operating assets and liabilities of $5.1 million. The significant items in the change in operating assets that impacted our use of cash in operations include a decrease of $9.3 million in accrued direct research and other current and long-term liabilities primarily due to the change in timing of bonus payments from December to March of the following year, which began with the December 2020 payments being delayed to March 2021, and to the termination of PAH-related studies and the completion of CARDINAL and MOXIe clinical trials, offset by an increase in accounts payable of $3.6 million due to timing of payments.

Investing Activities

Net cash used in operatinginvesting activities was $41.2$0.3 million and $0.2 million for the three months ended March 31, 2020, consisting primarily of a net loss of $48.9 million adjusted for non-cash items including stock-based compensation expense of $19.3 million, depreciation, amortization,2022 and imputed interest expense of $2.3 million, and a net increase in operating assets and liabilities of $13.9 million.  The significant items in the change in operating assets that impacted our use of cash in operations include increases in income tax receivable of $22.2 million and accounts payable of $10.7 million due to timing of payments, offset by a decrease in deferred revenue of $1.2 million due to the ratable recognition of revenue over the expected term of the performance obligation under the KKC Agreement, which resulted in recognition of $1.2 million of license and milestone revenue.

Investing Activities

Net cash used in investing activities was $0.2 million and $0.1 million for the three months ended March 31, 2021, and 2020, respectively, consisting of purchases of property and equipmentequipment..


Financing Activities

Net cash provided by financing activities was $4.7$0.2 million and $1.4$4.7 million for the three months ended March 31, 20212022 and 2020,2021, respectively, consisting of stock options exercised.exercises.

31


Operating Capital Requirements

To date, we have not generated any revenue from product sales. We do not know when or whether we will generate any revenue from product sales. We do not expect to generate significant revenue from product sales unless and until we obtain regulatory approval of and commercialize one or more of our current or future product candidates. We anticipate that we will continue to generate losses for the foreseeable future, and we expect the losses to increase as we continue the development of, and seek regulatory approvals for, our product candidates, and begin to commercialize any approved products. We are subject to all the risks related to the development and commercialization of novel therapeutics, including those described under the heading “Risk Factors” included in our most recent Annual Report on Form 10-K, and we may encounter unforeseen expenses, difficulties, complications, delays, and other unknown factors that may adversely affect our business. We continue to incur additional costs associated with operating as a public company. We anticipate that we will need substantial additional funding in connection with our continuing operations.

In October 2019, we entered into the 2019 Lease Agreement, relating to a new headquarter building lease of approximately 327,400 square feet of office and laboratory space located in Plano, Texas.

In December 2021, we obtained control of the building, and, accordingly, we recorded related right-of-use assets and the lease liabilities during the fourth quarter of 2021.
We have paused the tenant improvement activities for the new headquarter building and are attempting to sublease the building. At this point, we will not spend the earlier-planned $50 million in capital expenditures. If at a future date we determine to move into the building, capital expenditures will need to be incurred based on our occupancy requirements at that time.
The initial term of the lease is 16 years, with up to ten years of extension at our option. The annual base rent payment, which will begin in June 2022, will be determined based on the project cost, subject to an initial annual cap of approximately $13.3 million. Beginning in the third lease year, the base rent will increase 1.95% per annum each year. In addition to the annual base rent, we will pay for taxes, insurance, utilities, operating expenses, assessments under private covenants, maintenance and repairs, certain capital repairs and replacements, and building management fees.

In July 2021, Kyowa Kirin announced the submission of an NDA in Japan for bardoxolone for improvement of renal function in patients with Alport syndrome. We earned a $5.0 million milestone related to this event that was received and began to be recognized in the third quarter of 2021.

In December 2020, we closed a follow-on underwritten public offering of 2,000,000 shares of our Class A common stock for gross proceeds of $281.7 million. Net proceeds to us from the offering were approximately $277.5 million, after deducting underwriting discounts and commissions and offering expenses.

In October 2020, we entered into a lease agreement with the owner of our headquarters and offices located in Plano, Texas, with lease terms extending through June 30, 2022 and an option to renew up to three months.  We recorded approximately $4.8 million as a right-of-use asset and lease liability in October 2020.

In June 2020, we closed on the Development Agreement and Purchase Agreement, each dated June 10, 2020, under which certain BXLS entities paid us an aggregate of $350.0 million in exchange for future royalties on bardoxolone and an aggregate of 340,793 shares of our Class A common stock at $146.72 per share.

In June 2020, we paid off our Term Loans, which included payments for principal of $155.0 million, prepayment fees of $5.4 million, exit fees of $6.7 million, and accrued and unpaid interest of $1.0 million.

In March 2020, the United States enacted the CARES Act.  Under its provisions, we recognized a tax benefit and receivable of $22.2 million associated with the ability to carryback an applicable prior year’s net operating losses to a preceding year to generate a refund.  

In October 2019, we entered into the 2019 Lease Agreement, relating to the lease of approximately 327,400 square feet of office and laboratory space located in Plano, Texas.  The term of the lease is estimated to commence mid-2022, when construction is completed, and continue for 16 years, with up to 10 years of extension at our option.  The initial annual base rent will be determined based on the project cost, subject to an initial annual cap of approximately $13.3 million, which may increase in certain circumstances.  Beginning in the third lease year, the base rent will increase 1.95% per annum each year.  In addition to the annual base rent, we will pay for taxes, insurance, utilities, operating expenses, assessments under private covenants, maintenance and repairs, certain capital repairs and replacements, and building management fees.

In October 2019, we and AbbVie entered into the Reacquisition Agreement pursuant to which we reacquired the development, manufacturing, and commercialization rights concerning our proprietary Nrf2 activator product platform originally licensed to AbbVie in the AbbVie License Agreement and the Collaboration Agreement.  In exchange for such rights, we will pay AbbVie $330.0 million, of which total payments of $250.0 million have been made as of March 31, 2021, with the remaining $80.0 million payable on November 30, 2021.  We will also pay AbbVie an escalating, low single-digit royalty on worldwide net sales, on a product-by-product basis, of omaveloxolone and an identified list of certain next-generation Nrf2 activators.


Our longer term liquidity requirements will require us to raise additional capital, such as through additional equity, debt, or royalty financings or collaboration arrangements. Our future capital requirements will depend on many factors, including the receipt of milestones under our KKCKyowa Kirin Agreement and the timing of our expenditures related to clinical trials. We believe our existing cash and cash equivalents will be sufficient to enable us to fund our operations through mid-2024.the fourth quarter of 2024. However, we anticipate opportunistically raising additional capital before that time through equity offerings, collaboration or license agreements, additional debt financings, or royalty financings in order to maintain adequate capital reserves. In addition, we may choose to raise additional capital at any time for the further development of our existing product candidates and may also need to raise additional funds sooner to pursue other development activities related to additional product candidates. Decisions about the timing or nature of any financing will be based on, among other things, our perception of our liquidity and of the market opportunity to raise equity, debt, or royalty financing. Additional securities may include common stock, preferred stock, or debt securities. We may explore strategic collaborations or license arrangements for any of our product candidates. If we do explore any arrangements, there can be no assurance that any agreement will be reached, and we may determine to cease exploring a potential transaction for any or all of the assets at any time. If an agreement is reached, there can be no assurance that any such transaction would provide us with a material amount of additional capital resources.resources.

32


Until we can generate a sufficient amount of revenue from our product candidates, if ever, we expect to finance future cash needs through public or private equity or debt offerings, loans, royalty financings, and collaboration or license transactions. The outbreak of COVID-19 has caused significant disruption ofRecent and continued volatility in global financial markets which may reduce our ability to access capital, which could negatively affect our liquidity. Additional capital may not be available on reasonable terms, if at all. If we are unable to raise additional capital in sufficient amounts or on terms acceptable to us, we may have to significantly delay, scale back, or discontinue the development or commercialization of one or more of our product candidates. If we raise additional funds through the issuance of additional equity or debt securities, it could result in dilution to our existing stockholders or increased fixed payment obligations, and any such securities may have rights senior to those of our common stock. If we incur indebtedness or obtain royalty financing, we could become subject to covenants that would restrict our operations and potentially impair our competitiveness, such as limitations on our ability to incur additional debt, limitations on our ability to acquire, sell, or license intellectual property rights, and other operating restrictions that could adversely affect our ability to conduct our business, and any such debt or royalty financing could be secured by some or all of our assets. Any of these events could significantly harm our business, financial condition, and prospects.prospects. For a description of the numerous risks and uncertainties associated with product development and raising additional capital, see “Risk Factors” included in our Annual Report on Form 10-K for the year ended December 31, 2020.2021.

Our forecast of the period through which our financial resources will be adequate to support our operations is a forward-looking statement and involves risks and uncertainties, and actual results could vary as a result of a number of factors. We have based this estimate on assumptions that may prove to be wrong, and we could utilize our available capital resources sooner than we currently expect. Our future funding requirements, both near- and long-term, will depend on many factors, including, but not limited to:

the scope, rate of progress, results, and cost of our clinical trials, preclinical testing, and other activities related to the development of our product candidates;
the number and characteristics of product candidates that we pursue;
the costs of development efforts for our product candidates that are not subject to reimbursement from our collaborators;
the costs necessary to obtain regulatory approvals, if any, for our product candidates in the United States and other jurisdictions, and the costs of post-marketing studies that could be required by regulatory authorities in jurisdictions where approval is obtained;
the continuation of our existing collaboration with Kyowa Kirin and entry into new collaborations and the receipt of any collaboration payments;
the time and unreimbursed costs necessary to commercialize products in territories in which our product candidates are approved for sale;
the revenue from any future sales of our products or for which we are entitled to a profit share, royalties, and milestones;
the level of reimbursement or third-party payor pricing available to our products;
the costs of obtaining third-party commercial supplies of our products, if any, manufactured in accordance with regulatory requirements;
the costs associated with any potential loss or corruption of our information or data in a cyberattack on our computer systems or those of our suppliers, vendors, or collaborators who store or transmit our data;
the costs associated with being a public company;
any additional costs we incur, or delays in clinical trials we experience, associated with the COVID-19 pandemic; and
the costs we incur in the filing, prosecution, maintenance, and defense of our patent portfolio and other intellectual property rights.

the scope, rate of progress, results, and cost of our clinical trials, preclinical testing, and other activities related to the development of our product candidates;

the number and characteristics of product candidates that we pursue;

the costs of development efforts for our product candidates that are not subject to reimbursement from our collaborators;

the costs necessary to obtain regulatory approvals, if any, for our product candidates in the United States and other jurisdictions, and the costs of post-marketing studies that could be required by regulatory authorities in jurisdictions where approval is obtained;

the continuation of our existing collaboration with KKC and entry into new collaborations and the receipt of any collaboration payments;


the time and unreimbursed costs necessary to commercialize products in territories in which our product candidates are approved for sale;

the revenue from any future sales of our products for which we are entitled to a profit share, royalties, and milestones;

the level of reimbursement or third-party payor pricing available to our products;

the costs of obtaining third-party commercial supplies of our products, if any, manufactured in accordance with regulatory requirements;

the costs associated with any potential loss or corruption of our information or data in a cyberattack on our computer systems or those of our suppliers, vendors, or collaborators who store or transmit our data;

the costs associated with being a public company;

any additional costs we incur, or delays in clinical trials we experience, associated with the COVID-19 pandemic; and

the costs we incur in the filing, prosecution, maintenance, and defense of our patent portfolio and other intellectual property rights.

If we cannot expand our operations or otherwise capitalize on our business opportunities because we lack sufficient capital, our business, financial condition, and results of operations could be materially adversely affected.

33


Contractual Obligations and Commitments

We have various contractual obligations and other commitments that require payments at certain specified periods. The following table summarizes our contractual obligations and commitments as of March 31, 20212022 (unaudited):

 

 

Payments due by period

 

 

 

Less than

1 year

 

 

1 to 3

years

 

 

4 to 5

years

 

 

6 years and beyond

 

 

Total

 

 

 

(unaudited)

 

 

 

(in thousands)

 

Operating lease obligations(1)

 

$

2,907

 

 

$

14,271

 

 

$

26,927

 

 

$

169,478

 

 

$

213,583

 

Payable to collaborators

 

 

80,000

 

 

 

 

 

 

 

 

 

 

 

 

80,000

 

Total contractual obligations

 

$

82,907

 

 

$

14,271

 

 

$

26,927

 

 

$

169,478

 

 

$

293,583

 

 

 

Payments due by period

 

 

 

Less than
1 year

 

 

1 to 3
years

 

 

4 to 5
years

 

 

6 years and beyond

 

 

Total

 

 

 

(unaudited, in thousands)

 

Operating lease obligations(1)

 

$

12,633

 

 

$

17,448

 

 

$

27,831

 

 

$

179,687

 

 

$

237,599

 

Total contractual obligations

 

$

12,633

 

 

$

17,448

 

 

$

27,831

 

 

$

179,687

 

 

$

237,599

 

(1) Above table assumes one year rent abatement is applied beginning in June 2023 following FDA approval of omaveloxolone.

(1)

Operating lease obligations include current estimated payments for leases that have not yet commenced, net of lease incentives.

The terms of the Development Agreement require us to pay potential future royalty payments based on product development success. The above table excludes such obligations as the amount and timing of such obligations are unknown or uncertain, which are further described in Note 5, 4, Liability Related to Sale of Future Royalties, to Consolidated Financial Statements contained in this Quarterly Report on Form 10-Q.


Clinical Trials

As of March 31, 2021,2022, we have several on-going clinical trials in various stages. Under agreements with various CROs and clinical trial sites, we incur expenses related to clinical trials of our product candidates and potential other clinical candidates. The timing and amounts of these disbursements are contingent upon the achievement of certain milestones, patient enrollment, and services rendered or as expenses are incurred by the CROs or clinical trial sites. Therefore, we cannot estimate the potential timing and amount of these payments, and they have been excluded from the table above.

Critical Accounting Policies and Significant Judgments and Estimates

Our management’s discussion and analysis of our financial condition and results of operations are based on our consolidated financial statements, which have been prepared in accordance with U.S. GAAP. The preparation of these financial statements requires us to make estimates and judgments that affect the reported amounts of assets, liabilities, and expenses and the disclosure of contingent assets and liabilities in our financial statements. On an ongoing basis, we evaluate our estimates and judgments, including those related to revenue recognition, accrued research and development expenses, income taxes, and stock-based compensation. We base our estimates on historical experience, known trends and events, and various other factors that we believe to be reasonable under the circumstances, the results of which form the basis for making judgments about the carrying values of assets and liabilities that are not readily apparent from other sources. Actual results may differ from these estimates under different assumptions or conditions.

Our significant accounting policies are described in Note 2 of Part I, Item 1 of this Quarterly Report on Form 10-Q and in Part I, Item 7, “Critical Accounting Policies and Significant Judgments and Estimates” in our Annual Report on Form 10-K. There have been no changes to our critical accounting policies and estimates since our Annual Report on Form 10-K for the year ended December 31, 2020.2021.

Off-Balance Sheet Arrangements

Since our inception, we have not had any relationships with unconsolidated organizations or financial partnerships, such as structured finance or special purpose entities, that would have been established for the purpose of facilitating off-balance sheet arrangements, and we have not engaged in any other off-balance sheet arrangements, as defined in the rules and regulations of the SEC.

Recent Accounting Pronouncements

For a discussion of recent accounting pronouncements, please see Note 2, Summary of Significant Accounting Policies, of Notes to Consolidated Financial Statements contained in this Quarterly Report on Form 10-Q.

34


Item 3. Quantitative and Qualitative Disclosures About Market Risk.

We are exposed to market risks in the ordinary course of our business. These market risks are principally limited to interest rate fluctuations. We had cash and cash equivalents of $777.6$532.0 million at March 31, 2021,2022, consisting primarily of funds in operating cash accounts. The primary objective of our investment activities is to preserve principal and liquidity while maximizing income without significantly increasing risk. We do not enter into investments for trading or speculative purposes. Due to the short-term nature of our investment portfolio, we do not believe an immediate increase of 100 basis points in interest rates would have a material effect on the fair market value of our portfolio, and accordingly we do not expect a sudden change in market interest rates to affect materially our operating results or cash flows.

We contract with research, development, and manufacturing organizations and investigational sites globally. Generally, these contracts are denominated in United States dollars. However, we may be subject to fluctuations


in foreign currency rates in connection with agreements not denominated in United States dollars. We do not hedge our foreign currency exchange rate risk.

Item 4. Controls and Procedures.

Evaluation of Disclosure Controls and Procedures

Our management, with the participation of our Chief Executive Officer and Chief Financial Officer, evaluated the effectiveness of our disclosure controls and procedures as of March 31, 2021.2022. The term “disclosure controls and procedures,” as defined in Rules 13a-15(e) and 15d-15(e) under the Securities Exchange Act of 1934 (Exchange Act), means controls and other procedures of a company that are designed to ensure that information required to be disclosed by a company in the reports that it files or submits under the Exchange Act is recorded, processed, summarized, and reported, within the time periods specified in the SEC’s rules and forms. Disclosure controls and procedures include, without limitation, controls and procedures designed to ensure that information required to be disclosed by a company in the reports that it files or submits under the Exchange Act is accumulated and communicated to the company’s management, including its principal executive and principal financial officers, or persons performing similar functions, as appropriate to allow timely decisions regarding required disclosure. Management recognizes that any controls and procedures, no matter how well designed and operated, can provide only reasonable assurance of achieving their objectives, and management necessarily applies its judgment in evaluating the cost-benefit relationship of possible controls and procedures. Based on the evaluation of our disclosure controls and procedures as of March 31, 2021,2022, our Chief Executive Officer and Chief Financial Officer concluded that, as of such date, our disclosure controls and procedures were effective at the reasonable assurance level.

Changes in Internal Control Over Financial Reporting

There have been no changes in our internal control over financial reporting, as such term is defined in Rules 13a-15(f) and 15d-15(f) promulgated under the Exchange Act, during the three months ended March 31, 2021,2022, that have materially affected, or are reasonably likely to materially affect, our internal control over financial reporting.

35


PART II — OTHER INFORMATION

For a discussion of material pending legal proceedings, please read Note 11,10, Commitments and Contingencies – Litigation, to our condensed consolidated financial statements included in Part I, Item I, “Financial Statements (Unaudited),” of this Quarterly Report on Form 10-Q, which is incorporated into this item by reference.

Item 1A. Risk Factors.

In addition to other information set forth in this Quarterly Report on Form 10-Q, you should carefully consider the risk factors and other cautionary statements described under the heading “Risk Factors” included in our Annual Report on Form 10-K for the year ended December 31, 2020,2021, which could materially affect our businesses, financial condition, or future results. Additional risks and uncertainties currently unknown to us, or that we currently deem to be immaterial, also may materially adversely affect our business, financial condition, or future results. There have been no material changes in our risk factors from those described in the Annual Report on Form 10-K for the year ended December 31, 2020.2021.

Item 2. Unregistered Sales of Equity Securities and Use of Proceeds.

None.


Item 3. Defaults Upon Senior Securities.

None.

Item 4. Mine Safety Disclosures.

None.

Item 5. Other Information.

None.


36


Item 6. Exhibits.

 

Exhibit

Number

 

Description

  3.1

Thirteenth Amended and Restated Certificate of Incorporation, dated May 11, 2016 (incorporated by reference to Exhibit 3.7 to the Company’s Form S-1 (File No. 333-208843), filed with the SEC on May 16, 2016).

  3.2

Second Amended and Restated Bylaws, dated as of December 7, 2016 (incorporated by reference to Exhibit 3.1 to the Company’s Form 8-K (File No. 001-37785), filed with the SEC on December 7, 2016).

  10.1+10.1*#

IndemnificationAmendment No. 1 to Exclusive License Agreement, dated as of April 5, 2022, by and between the CompanyKU Center for Technology Commercialization, Inc. and Christy J. Oliger,the Registrant, dated as of, April 15, 2021.as amended. (incorporated by reference to Exhibit 10.1 to the Company’s Form 8-K (File No. 001-37785), filed with the SEC on April 15, 2021).

  10.2+10.2*#

IndemnificationSeventh Supplement to Exclusive License and Supply Agreement, by and between the Company and Shamim Ruff, dated as of April 15, 2021. (incorporated by reference to Exhibit 10.2 to the Company’s Form 8-K (File No. 001-37785)February 28, 2022 between Reata Pharmaceuticals, Inc. and Kyowa Hakko Kirin Co., filed with the SEC on April 15, 2021).Ltd.

 10.3+

Notice of Grant of Restricted Stock Units for employees, dated as of December 2, 2020.

  31.1*

 

Certification of Principal Executive Officer Pursuant to Rules 13a-14(a) and 15d-14(a) under the Securities Exchange Act of 1934, as Adopted Pursuant to Section 302 of the Sarbanes-Oxley Act of 2002.

  31.2*

 

Certification of Principal Financial Officer Pursuant to Rules 13a-14(a) and 15d-14(a) under the Securities Exchange Act of 1934, as Adopted Pursuant to Section 302 of the Sarbanes-Oxley Act of 2002.

  32.1**

 

Certification of Principal Executive Officer Pursuant to 18 U.S.C. Section 1350, as Adopted Pursuant to Section 906 of the Sarbanes-Oxley Act of 2002.

  32.2**

 

Certification of Principal Financial Officer Pursuant to 18 U.S.C. Section 1350, as Adopted Pursuant to Section 906 of the Sarbanes-Oxley Act of 2002.

101.INS*

 

Inline XBRL Instance Document - The cover page interactive data file does not appear in the interactive data file because its XBRL tags are embedded within the inline XBRL document

101.SCH*

Inline XBRL Taxonomy Extension Schema Document

101.CAL*

Inline XBRL Taxonomy Extension Calculation Linkbase Document

101.DEF*

Inline XBRL Taxonomy Extension Definition Linkbase Document

101.LAB*

Inline XBRL Taxonomy Extension Label Linkbase Document

101.PRE*

Inline XBRL Taxonomy Extension Presentation Linkbase Document

104

Cover Page Interactive Data File (embedded within the Inline XBRL document)

*

Filed herewith.

**

Furnished herewith.

* Filed herewith.

** Furnished herewith.

# Information in this exhibit identified by three asterisks [***] is confidential and has been omitted pursuant to

Item 601(b)(10)(iv) of Regulation S-K because it is not material and is the type of information that the Company customarily treats as private or confidential. An unredacted copy of this exhibit will be furnished to the Securities and Exchange Commission on a supplemental basis upon request.

37


SIGNATURES

+

Indicates management contract or compensatory plan.


SIGNATURES

Pursuant to the requirements of the Securities Exchange Act of 1934, the registrant has duly caused this report to be signed on its behalf by the undersigned, thereunto duly authorized.

 

Date: May 6, 202110, 2022

REATA PHARMACEUTICALS, INC.

 

 

 

By:

/s/ J. Warren Huff

 

Name:

J. Warren Huff

 

Title:

Chief Executive Officer and President

 

By:

/s/ Manmeet S. Soni

 

Name:

Manmeet S. Soni

 

Title:

Chief Operating Officer, Chief Financial Officer, and Executive Vice President

38