SS

UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

Washington, D.C. 20549

 

FORM 10-Q

 

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

For the quarterly period ended March 31, 20232024

or

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: 001-37923

 

CRISPR THERAPEUTICS AG

(Exact name of registrant as specified in its charter)

 

 

Switzerland

Not Applicable

(State or other jurisdiction of

incorporation or organization)

(I.R.S. Employer

Identification No.)

 

 

Baarerstrasse 14

6300 Zug, Switzerland

Not Applicable

(Address of principal executive offices)

(Zip Code)

+41 (0)41 561 32 77

(Registrant’s telephone number, including area code)

 

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

 

Title of each class

Trading Symbol(s)

Name of each exchange on which registered

Common Shares, nominal value CHF 0.03

CRSP

The 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. YesNo

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). YesNo

Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, a smaller reporting company, or emerging growth company. See definition 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 registrant is a shell company (as defined in Rule 12b-2 of the Exchange Act). Yes ☐ No

As of May 5, 2023,3, 2024, there were 78,935,09884,918,093 shares of registrant’s common shares outstanding.

 


Throughout this Quarterly Report on Form 10-Q, the “Company,” “CRISPR,” “CRISPR Therapeutics,” “we,” “us,” and “our,” except where the context requires otherwise, refer to CRISPR Therapeutics AG and its consolidated subsidiaries.subsidiaries;“our board of directors” refers to the board of directors of CRISPR Therapeutics AG; and we generally refer to CASGEVY (exagamglogene autotemcel [exa-cel]), formerly CTX001, as "CASGEVY".

“CRISPR Therapeutics®Therapeutics®” standard character mark and design logo, “COBALTTM,”“CRISPRXTM,“CRISPRXTM,” “CRISPR TXTM,TXTM,“CTX001TM,“CTX112TM,“CTX110®,“CTX131TM,“CTX112TM,“CTX310TM,“CTX130TM,“CTX320TM,“CTX131TM,”“CTX310TM,”“CTX320TM,”“VCTX210TM“CTX330TM,” and “VCTX211TM,” “CTX211TM” are trademarks and registered trademarks of CRISPR Therapeutics AG. The CASGEVY™ word mark and design are trademarks of Vertex Pharmaceuticals Incorporated. All other trademarks and registered trademarks contained in this Quarterly Report on Form 10-Q are the property of their respective owners. Solely for convenience, trademarks, service marks and trade names referred to in this Quarterly Report on Form 10-Q may appear without the ® or symbols and any such omission is not intended to indicate waiver of any such rights.

 

FORWARD-LOOKING STATEMENTS

This Quarterly Report on Form 10-Q contains “forward-looking statements” that involve substantial risks and uncertainties. All statements, other than statements of historical facts, contained in this Quarterly Report on Form 10-Q are forward-looking statements. These statements are often identified by the use of words such as “anticipate,” “believe,” “continue,” “could,” “estimate,” “expect,” “intend,” “may,” “plan,” “predict,” “project,” “potential,” “will,” “would” or the negative or plural of these words or similar expressions or variations, although not all forward-looking statements contain these identifying words. Forward-looking statements in this Quarterly Report on Form 10-Q include, but are not limited to, statements about:

the safety, efficacy and clinical progress of our various clinical programs, including those for exa-cel (formerly knownCASGEVY, CTX112, CTX131, CTX211, CTX310 and CTX320;
plans and expectations for the commercialization of, and anticipated benefits of, CASGEVY, including the anticipated patient population eligible for CASGEVY in the United States and plans for patient access to CASGEVY, as CTX001), CTX110, CTX112, CTX130, CTX131, VCTX210well as timelines for and VCTX211;expectations regarding additional regulatory agency decisions regarding CASGEVY;
the status of clinical trials, including development timelines regulatory submissions and discussions with regulatory authorities related to product candidates under development by us and our collaborators;
the initiation, timing, progress and results of our preclinical studies and clinical trials, including our ongoing clinical trials and any planned clinical trials, and our research and development programs, including delays or disruptions in clinical trials, non-clinical experiments and Investigational New Drug, or IND, application-enabling studies;
the actual or potential benefits of U.S. Food and Drug Administration, or FDA,regulatory designations, such as Orphan Drug, Fast Track and regenerative medicine advanced therapy, or RMAT, in the United States or such European equivalents, including PRIority MEdicines, or PRIME, designation;
our ability to advance product candidates into, and successfully complete, clinical trials;
the size and growth potential of the markets for our product candidates and our ability to serve those markets;
the rate and degree of market acceptance of our product candidates and the success of competing therapies that are or become available;
our efforts to establish internal manufacturing capabilities and operation of our cell therapy manufacturing facility;
our intellectual property coverage and positions, including those of our licensors and third parties as well as the status and potential outcome of proceedings involving any such intellectual property;
our anticipated expenses, ability to obtain funding for our operations and the sufficiency of our cash resources; and
the therapeutic value, development, and commercial potential of CRISPR/Cas9 gene editing technologies and therapies.

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 assumptions that could cause our actual results and the timing of certain events to differ materially from future results expressed or implied by the forward-looking statements. Factors that could cause or contribute to such differences include, but are not limited to, those identified herein, and those discussed in the section titled “Risk Factors,” set forth in Part II, Item 1A of this Quarterly Report on Form 10-Q, if any, our Annual Report on Form 10-K filed with the Securities and Exchange Commission, or SEC, on February 21, 2023,2024, and in other SEC filings. You should not rely upon forward-looking statements as predictions of future events. Such forward-looking statements speak only as of the date of this report. Our forward-looking statements do not reflect the potential impact of any future acquisitions, mergers, dispositions, joint ventures or investments we may make or enter into.

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, performance or achievements may be materially different from what we expect. Except as required by law, we undertake no obligation to update any forward-looking statements to reflect events or circumstances after the date of such statements.


Investors and others should note that we announce material information to our investors using our investor relations website (https://crisprtx.gcs-web.com/), SEC filings, press releases, public conference calls and webcasts. We use these channels as well as social media to communicate with the public about our Company, our business, our product candidates and other matters. It is possible that the information we post on social media could be deemed to be material information. Therefore, we encourage investors, the media, and others interested in our Company to review the information we post on the social media channels listed on our investor relations website.


Index

 

Page

Number

PART I: FINANCIAL INFORMATION

 

 

 

Item 1. Condensed Consolidated Financial Statements (unaudited)

2

 

 

Condensed Consolidated Balance Sheets as of March 31, 20232024 and December 31, 20222023

2

 

 

Condensed Consolidated Statements of Operations and Comprehensive Loss for the three months ended March 31, 20232024 and 20222023

3

 

 

Condensed Consolidated Statements of Shareholders’ Equity for the three months ended March 31, 20232024 and 20222023

4

 

 

Condensed Consolidated Statements of Cash Flows for the three months ended March 31, 20232024 and 20222023

5

 

 

Notes to Condensed Consolidated Financial Statements

6

 

 

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

1617

 

 

Item 3. Quantitative and Qualitative Disclosures about Market Risk

2425

 

 

Item 4. Controls and Procedures

25

 

 

PART II: OTHER INFORMATION

 

 

 

Item 1. Legal Proceedings

26

 

 

Item 1A. Risk Factors

26

 

 

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

26

 

 

Item 3. Defaults Upon Senior Securities

26

 

 

Item 4. Mine Safety Disclosures

26

 

 

Item 5. Other Information

2726

 

 

Item 6. Exhibits

27

 

 

SIGNATURES

28

 


 


PART I—FINANCIAL INFORMATION

Item 1. Financial Statements

CRISPR Therapeutics AG

Condensed Consolidated Balance Sheets

(unaudited, in thousands, except share and per share data)

 

 

As of

 

 

As of

 

 

March 31,

 

December 31,

 

 

March 31,

 

December 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Assets

Assets

 

Assets

 

Current assets:

 

 

 

 

 

 

 

 

 

 

 

 

Cash and cash equivalents

 

$

344,407

 

 

$

211,885

 

 

$

707,427

 

 

$

389,477

 

Marketable securities

 

 

1,538,763

 

 

 

1,603,433

 

 

 

1,400,698

 

 

 

1,304,215

 

Accounts receivable

 

 

 

 

 

200,000

 

Prepaid expenses and other current assets

 

 

24,796

 

 

 

37,708

 

 

 

11,499

 

 

 

14,386

 

Total current assets

 

 

1,907,966

 

 

 

1,853,026

 

 

 

2,119,624

 

 

 

1,908,078

 

Property and equipment, net

 

 

162,198

 

 

 

163,634

 

 

 

147,253

 

 

 

151,945

 

Marketable securities, non-current

 

 

6,320

 

 

 

53,130

 

 

 

 

 

 

1,973

 

Intangible assets, net

 

 

57

 

 

 

71

 

 

 

2

 

 

 

16

 

Restricted cash

 

 

11,717

 

 

 

11,635

 

 

 

11,516

 

 

 

11,591

 

Operating lease assets

 

 

153,346

 

 

 

156,921

 

 

 

151,254

 

 

 

153,993

 

Other non-current assets

 

 

2,760

 

 

 

4,640

 

 

 

9,368

 

 

 

1,975

 

Total assets

 

$

2,244,364

 

 

$

2,243,057

 

 

$

2,439,017

 

 

$

2,229,571

 

Liabilities and shareholders’ equity

Liabilities and shareholders’ equity

 

Liabilities and shareholders’ equity

 

Current liabilities:

 

 

 

 

 

 

 

 

 

 

 

 

Accounts payable

 

$

45,153

 

 

$

27,428

 

 

$

16,463

 

 

$

38,147

 

Accrued expenses

 

 

85,631

 

 

 

77,682

 

 

 

76,432

 

 

 

45,335

 

Deferred revenue, current

 

 

4,072

 

 

 

4,105

 

Accrued tax liabilities

 

 

273

 

 

 

135

 

 

 

1,207

 

 

 

438

 

Operating lease liabilities

 

 

15,717

 

 

 

15,842

 

 

 

15,992

 

 

 

15,625

 

Other current liabilities

 

 

20

 

 

 

20

 

 

 

4,824

 

 

 

5,141

 

Total current liabilities

 

 

146,794

 

 

 

121,107

 

 

 

118,990

 

 

 

108,791

 

Deferred revenue, non-current

 

 

12,323

 

 

 

12,323

 

 

 

13,541

 

 

 

14,012

 

Operating lease liabilities, net of current portion

 

 

224,552

 

 

 

228,179

 

 

 

218,935

 

 

 

223,007

 

Other non-current liabilities

 

 

5,799

 

 

 

5,969

 

 

 

3,615

 

 

 

958

 

Total liabilities

 

 

389,468

 

 

 

367,578

 

 

 

355,081

 

 

 

346,768

 

Commitments and contingencies, see Note 7

 

 

 

 

 

 

 

 

 

 

 

 

Shareholders’ equity:

 

 

 

 

 

 

 

 

 

 

 

 

Common shares, CHF 0.03 par value, 150,347,467 shares authorized at
March 31, 2023 and at December 31, 2022,
79,044,050 and 78,692,766
shares issued at March 31, 2023 and December 31, 2022, respectively,
78,863,734
and
78,512,450 shares outstanding at March 31, 2023 and December 31, 2022,
respectively

 

 

2,452

 

 

 

2,441

 

Treasury shares, at cost, 180,316 shares at March 31, 2023 and at December 31, 2022

 

 

(63

)

 

 

(63

)

Common shares, CHF 0.03 nominal value, 130,452,265 and 126,536,183 shares authorized at
March 31, 2024 and December 31, 2023, respectively,
85,007,926 and 80,214,694 shares issued at March 31, 2024 and December 31, 2023, respectively, 84,837,610 and 80,044,378 shares outstanding at March 31, 2024 and December 31, 2023, respectively

 

 

2,658

 

 

 

2,497

 

Treasury shares, at cost, 170,316 shares at March 31, 2024 and at December 31, 2023

 

 

(62

)

 

 

(62

)

Additional paid-in capital

 

 

2,761,050

 

 

 

2,734,838

 

 

 

3,199,183

 

 

 

2,878,155

 

Accumulated deficit

 

 

(899,155

)

 

 

(846,090

)

 

 

(1,116,291

)

 

 

(999,700

)

Accumulated other comprehensive loss

 

 

(9,388

)

 

 

(15,647

)

Accumulated other comprehensive (loss) income

 

 

(1,552

)

 

 

1,913

 

Total shareholders' equity

 

 

1,854,896

 

 

 

1,875,479

 

 

 

2,083,936

 

 

 

1,882,803

 

Total liabilities and shareholders’ equity

 

$

2,244,364

 

 

$

2,243,057

 

 

$

2,439,017

 

 

$

2,229,571

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

2


 

CRISPR Therapeutics AG

Condensed Consolidated Statements of Operations and Comprehensive Loss

(unaudited, in thousands, except share and per share data)

 

 

Three Months Ended

 

 

Three Months Ended

 

 

March 31,

 

 

March 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Revenue:

 

 

 

 

 

 

 

 

 

 

 

 

Collaboration revenue

 

$

100,000

 

 

$

178

 

 

$

 

 

$

100,000

 

Grant revenue

 

 

 

 

 

762

 

 

 

504

 

 

 

 

Total revenue

 

 

100,000

 

 

 

940

 

 

 

504

 

 

 

100,000

 

Operating expenses:

 

 

 

 

 

 

 

 

 

 

 

 

Research and development

 

 

99,935

 

 

 

118,245

 

 

 

76,172

 

 

 

99,935

 

General and administrative

 

 

22,360

 

 

 

28,021

 

 

 

17,953

 

 

 

22,360

 

Collaboration expense, net

 

 

42,192

 

 

 

30,646

 

 

 

46,966

 

 

 

42,192

 

Total operating expenses

 

 

164,487

 

 

 

176,912

 

 

 

141,091

 

 

 

164,487

 

Loss from operations

 

 

(64,487

)

 

 

(175,972

)

 

 

(140,587

)

 

 

(64,487

)

Other income:

 

 

 

 

 

 

 

 

 

 

 

 

Other income, net

 

 

12,742

 

 

 

363

 

 

 

24,720

 

 

 

12,742

 

Total other income, net

 

 

12,742

 

 

 

363

 

 

 

24,720

 

 

 

12,742

 

Net loss before income taxes

 

 

(51,745

)

 

 

(175,609

)

 

 

(115,867

)

 

 

(51,745

)

Provision for income taxes

 

 

(1,320

)

 

 

(3,608

)

 

 

(724

)

 

 

(1,320

)

Net loss

 

 

(53,065

)

 

 

(179,217

)

 

 

(116,591

)

 

 

(53,065

)

Foreign currency translation adjustment

 

 

32

 

 

 

(27

)

 

 

(11

)

 

 

32

 

Unrealized gain (loss) on marketable securities

 

 

6,227

 

 

 

(11,799

)

Unrealized (loss) gain on marketable securities

 

 

(3,454

)

 

 

6,227

 

Comprehensive loss

 

$

(46,806

)

 

$

(191,043

)

 

$

(120,056

)

 

$

(46,806

)

 

 

 

 

 

 

 

 

 

 

 

 

Net loss per common share — basic

 

$

(0.67

)

 

$

(2.32

)

 

$

(1.43

)

 

$

(0.67

)

Basic weighted-average common shares outstanding

 

 

78,676,986

 

 

 

77,098,319

 

 

 

81,794,630

 

 

 

78,676,986

 

Net loss per common share — diluted

 

$

(0.67

)

 

$

(2.32

)

 

$

(1.43

)

 

$

(0.67

)

Diluted weighted-average common shares outstanding

 

 

78,676,986

 

 

 

77,098,319

 

 

 

81,794,630

 

 

 

78,676,986

 

 

 

 

 

 

 

 

 

 

 

 

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

3


 

CRISPR Therapeutics AG

Condensed Consolidated Statements of Shareholders’ Equity

(unaudited, in thousands, except share and per share data)

.

 

Common Shares

 

Treasury Shares

 

 

 

 

 

 

 

 

 

 

Shares

 

CHF 0.03
Par Value

 

Shares

 

Amount,
at cost

 

Additional
Paid-in
Capital

 

Accumulated
Deficit

 

Accumulated
Other
Comprehensive
Loss

 

Total
Shareholders’
Equity

 

Balance at December 31, 2021

 

76,990,066

 

$

2,391

 

 

180,316

 

$

(63

)

$

2,598,114

 

$

(195,915

)

$

(5,067

)

$

2,399,460

 

Vesting of restricted shares

 

123,564

 

 

4

 

 

 

 

 

 

 

 

 

 

 

 

4

 

Exercise of vested options, net of issuance costs of $0.2 million

 

261,280

 

 

12

 

 

 

 

 

 

9,998

 

 

 

 

 

 

10,010

 

Purchase of common stock under ESPP

 

11,495

 

 

 

 

 

 

 

 

740

 

 

 

 

 

 

740

 

Stock-based compensation expense

 

 

 

 

 

 

 

 

 

25,745

 

 

 

 

 

 

25,745

 

Other comprehensive loss

 

 

 

 

 

 

 

 

 

 

 

 

 

(11,826

)

 

(11,826

)

Net loss

 

 

 

 

 

 

 

 

 

 

 

(179,217

)

 

 

 

(179,217

)

Balance at March 31, 2022

 

77,386,405

 

$

2,407

 

 

180,316

 

$

(63

)

$

2,634,597

 

$

(375,132

)

$

(16,893

)

$

2,244,916

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Balance at December 31, 2022

 

78,512,450

 

$

2,441

 

 

180,316

 

$

(63

)

$

2,734,838

 

$

(846,090

)

$

(15,647

)

$

1,875,479

 

Vesting of restricted shares

 

172,995

 

 

5

 

 

 

 

 

 

 

 

 

 

 

 

5

 

Exercise of vested options, net of issuance costs of $0.2 million

 

159,184

 

 

6

 

 

 

 

 

 

4,677

 

 

 

 

 

 

4,683

 

Purchase of common stock under ESPP

 

19,105

 

 

 

 

 

 

 

 

660

 

 

 

 

 

 

660

 

Stock-based compensation expense

 

 

 

 

 

 

 

 

 

20,875

 

 

 

 

 

 

20,875

 

Other comprehensive income

 

 

 

 

 

 

 

 

 

 

 

 

 

6,259

 

 

6,259

 

Net loss

 

 

 

 

 

 

 

 

 

 

 

(53,065

)

 

 

 

(53,065

)

Balance at March 31, 2023

 

78,863,734

 

$

2,452

 

 

180,316

 

$

(63

)

$

2,761,050

 

$

(899,155

)

$

(9,388

)

$

1,854,896

 

 

Common Shares

 

Treasury Shares

 

 

 

 

 

 

 

 

 

 

Shares

 

CHF 0.03
Nominal Value

 

Shares

 

Amount,
at cost

 

Additional
Paid-in
Capital

 

Accumulated
Deficit

 

Accumulated
Other
Comprehensive
Gain/(Loss)

 

Total
Shareholders’
Equity

 

Balance at December 31, 2022

 

78,512,450

 

$

2,441

 

 

180,316

 

$

(63

)

$

2,734,838

 

$

(846,090

)

$

(15,647

)

$

1,875,479

 

Vesting of restricted shares

 

172,995

 

 

5

 

 

 

 

 

 

 

 

 

 

 

 

5

 

Exercise of vested options, net of issuance costs of $0.2 million

 

159,184

 

 

6

 

 

 

 

 

 

4,677

 

 

 

 

 

 

4,683

 

Purchase of common stock under ESPP

 

19,105

 

 

 

 

 

 

 

 

660

 

 

 

 

 

 

660

 

Stock-based compensation expense

 

 

 

 

 

 

 

 

 

20,875

 

 

 

 

 

 

20,875

 

Other comprehensive income

 

 

 

 

 

 

 

 

 

 

 

 

 

6,259

 

 

6,259

 

Net loss

 

 

 

 

 

 

 

 

 

 

 

(53,065

)

 

 

 

(53,065

)

Balance at March 31, 2023

 

78,863,734

 

$

2,452

 

 

180,316

 

$

(63

)

$

2,761,050

 

$

(899,155

)

$

(9,388

)

$

1,854,896

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Balance at December 31, 2023

 

80,044,378

 

$

2,497

 

 

170,316

 

$

(62

)

$

2,878,155

 

$

(999,700

)

$

1,913

 

$

1,882,803

 

Issuance of common stock, net of issuance costs of $3.8 million

 

3,929,610

 

 

132

 

 

 

 

 

 

277,015

 

 

 

 

 

 

277,147

 

Vesting of restricted shares

 

214,913

 

 

7

 

 

 

 

 

 

 

 

 

 

 

 

7

 

Exercise of vested options, net of issuance costs of $0.6 million

 

632,683

 

 

22

 

 

 

 

 

 

23,844

 

 

 

 

 

 

23,866

 

Purchase of common stock under ESPP

 

16,026

 

 

 

 

 

 

 

 

764

 

 

 

 

 

 

764

 

Stock-based compensation expense

 

 

 

 

 

 

 

 

 

19,405

 

 

 

 

 

 

19,405

 

Other comprehensive loss

 

 

 

 

 

 

 

 

 

 

 

 

 

(3,465

)

 

(3,465

)

Net loss

 

 

 

 

 

 

 

 

 

 

 

(116,591

)

 

 

 

(116,591

)

Balance at March 31, 2024

 

84,837,610

 

$

2,658

 

 

170,316

 

$

(62

)

$

3,199,183

 

$

(1,116,291

)

$

(1,552

)

$

2,083,936

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

4


 

CRISPR Therapeutics AG

Condensed Consolidated Statements of Cash Flows

(unaudited, in thousands)

 

 

Three Months Ended March 31,

 

 

Three Months Ended March 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Operating activities:

 

 

 

 

 

 

 

 

 

 

 

 

Net loss

 

$

(53,065

)

 

$

(179,217

)

 

$

(116,591

)

 

$

(53,065

)

Reconciliation of net loss to net cash used in operating activities:

 

 

 

 

 

 

 

 

 

 

Depreciation and amortization

 

 

5,051

 

 

 

6,039

 

 

 

4,833

 

 

 

5,051

 

Equity-based compensation

 

 

20,875

 

 

 

25,745

 

 

 

19,405

 

 

 

20,875

 

Other non-cash items, net

 

 

(969

)

 

 

6,099

 

 

 

(8,385

)

 

 

(969

)

Changes in:

 

 

 

 

 

 

 

 

 

 

Accounts receivable

 

 

 

 

 

162

 

 

 

200,000

 

 

 

 

Prepaid expenses and other assets

 

 

12,040

 

 

 

4,611

 

 

 

2,949

 

 

 

12,040

 

Accounts payable and accrued expenses

 

 

25,214

 

 

 

(1,616

)

 

 

9,443

 

 

 

25,214

 

Deferred revenue

 

 

 

 

 

(762

)

 

 

(504

)

 

 

 

Operating lease assets and liabilities

 

 

(177

)

 

 

3,854

 

 

 

(966

)

 

 

(177

)

Other liabilities, net

 

 

(170

)

 

 

(154

)

 

 

(432

)

 

 

(170

)

Net cash provided by (used in) operating activities

 

 

8,799

 

 

 

(135,239

)

Net cash provided by operating activities

 

 

109,752

 

 

 

8,799

 

Investing activities:

 

 

 

 

 

 

 

 

 

 

Purchase of property, plant and equipment

 

 

(3,059

)

 

 

(15,350

)

 

 

(721

)

 

 

(3,059

)

Investment in equity securities

 

 

(7,500

)

 

 

 

Purchases of marketable securities

 

 

(265,627

)

 

 

(323,140

)

 

 

(326,915

)

 

 

(265,627

)

Maturities of marketable securities

 

 

386,515

 

 

 

223,975

 

 

 

237,336

 

 

 

386,515

 

Net cash provided by (used in) investing activities

 

 

117,829

 

 

 

(114,515

)

Net cash (used in) provided by investing activities

 

 

(97,800

)

 

 

117,829

 

Financing activities:

 

 

 

 

 

 

 

 

 

 

Proceeds from issuance of common shares, net of issuance costs

 

 

280,745

 

 

 

 

Proceeds from exercise of options and ESPP contributions, net of issuance costs

 

 

5,404

 

 

 

10,649

 

 

 

25,189

 

 

 

5,404

 

Net cash provided by financing activities

 

 

5,404

 

 

 

10,649

 

 

 

305,934

 

 

 

5,404

 

Effect of exchange rate changes on cash

 

 

32

 

 

 

(27

)

 

 

(11

)

 

 

32

 

Increase (decrease) in cash

 

 

132,064

 

 

 

(239,132

)

Increase in cash

 

 

317,875

 

 

 

132,064

 

Cash, cash equivalents and restricted cash, beginning of period

 

 

224,060

 

 

 

939,944

 

 

 

401,068

 

 

 

224,060

 

Cash, cash equivalents and restricted cash, end of period

 

$

356,124

 

 

$

700,812

 

 

$

718,943

 

 

$

356,124

 

Supplemental disclosure of non-cash investing and financing activities

 

 

 

 

 

 

 

 

 

 

Property and equipment purchases in accounts payable and accrued expenses

 

$

2,663

 

 

$

5,888

 

 

$

131

 

 

$

2,633

 

Equity issuance costs in accounts payable and accrued expenses

 

$

155

 

 

$

217

 

Leasehold improvements paid directly by landlord

 

$

 

 

$

13,015

 

Equity issuance costs in accounts payable, accrued expenses, and other long-term liabilities

 

$

4,522

 

 

$

155

 

 

 

As of March 31,

 

 

As of March 31,

 

Reconciliation to amounts within the condensed consolidated balance sheets

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Cash and cash equivalents

 

 

344,407

 

 

 

683,906

 

 

$

707,427

 

 

$

344,407

 

Prepaid expenses and other current assets

 

 

 

 

 

4,783

 

Restricted cash

 

 

11,717

 

 

 

12,123

 

 

 

11,516

 

 

 

11,717

 

Cash, cash equivalents and restricted cash at end of period

 

$

356,124

 

 

$

700,812

 

 

$

718,943

 

 

$

356,124

 

 

The accompanying notes are an integral part of these unaudited condensed consolidated financial statements.

5


 

CRISPR Therapeutics AG

Notes to Condensed Consolidated Financial Statements

(unaudited)

1. Basis of Presentation and Significant Accounting Policies

Basis of Presentation

The accompanying condensed consolidated financial statements are unaudited and have been prepared by the Company in accordance with accounting principles generally accepted in the United States of America, or U.S. GAAP.

The accompanying condensed consolidated financial statements include the accounts of the Company and its wholly-owned subsidiaries. All intercompany balances and transactions have been eliminated in consolidation. The Company views its operations and manages its business in one operating segment, which is the business of discovering, developing and commercializing therapies derived from or incorporating genome-editing technology. Certain information and footnote disclosures normally included in the Company’s annual financial statements have been condensed or omitted. These interim financial statements, in the opinion of management, reflect all normal recurring adjustments necessary for a fair presentation of the financial position and results of operations for the three-month interim periods ended March 31, 20232024 and 2022.2023.

Beginning in 2022, "collaboration expense, net" in the condensed consolidated statements of operations and comprehensive loss present collaboration costs related to the exagamglogene autotemcel, or exa-cel (formerly known as CTX001), program under the Company's collaboration with Vertex Pharmaceuticals Incorporated and certain of its subsidiaries, or Vertex, accounted for under ASC 808, Collaborative Agreements, or ASC 808. Refer to Note 6 to these condensed consolidated financial statements for further discussion on the Company's agreements with Vertex, including the exa-cel program.

The results of operations for the interim periods are not necessarily indicative of the results of operations to be expected for the full year. These interim financial statements should be read in conjunction with the audited financial statements for the year ended December 31, 2022,2023, which are contained in the 20222023 Annual Report on Form 10-K filed with the Securities and Exchange Commission, or the SEC, on February 21, 2023.2024.

Use of Estimates

The preparation of financial statements in conformity with U.S. GAAP requires management to make estimates and assumptions that affect the amounts reported in the financial statements and accompanying notes. On an ongoing basis, the Company’s management evaluates its estimates, which include, but are not limited to, revenue recognition, equity-based compensation expense and reported amounts of expenses during the period. Significant estimates in these consolidated financial statements have been made in connection with revenue recognition and equity-based compensation expense. The Company bases its estimates on historical experience and other market-specific or other relevant assumptions that it believes to be reasonable under the circumstances. Actual results may differ from those estimates or assumptions. Changes in estimates are reflected in reported results in the period in which they become known.

Significant Accounting Policies

The significant accounting policies used in preparation of these condensed consolidated financial statements for the three months ended March 31, 20232024 are consistent with those discussed in Note 2 to the consolidated financial statements in the Company’s 20222023 Annual Report on Form 10-K filed with the SEC on February 21, 2023.2024.

New Accounting Pronouncements – Recently Adopted

From time to time, new accounting pronouncements are issued by the Financial Accounting Standards Board, or FASB, or other standard setting bodies that the Company adopts as of the specified effective date. The Company does not believe that the adoption of recently issued standards havehas or may have a material impact on its condensed consolidated financial statements and disclosures.

 

 

6


 

2. Marketable Securities

The following table summarizes cash equivalents and marketable securities held at March 31, 20232024 and December 31, 20222023 (in thousands), which are recorded at fair value. The table below excludes $263.6455.0 million and $159.3197.8 million of cash at March 31, 20232024 and December 31, 2022,2023, respectively.

 

 

Amortized
Cost

 

 

Gross
Unrealized
Gains

 

 

Gross
Unrealized
Losses

 

 

Fair Value

 

 

Amortized
Cost

 

 

Gross
Unrealized
Gains

 

 

Gross
Unrealized
Losses

 

 

Fair Value

 

March 31, 2023

 

 

 

 

 

 

 

 

 

 

 

 

March 31, 2024

 

 

 

 

 

 

 

 

 

 

 

 

Cash equivalents:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Money market funds

 

$

19,387

 

 

$

 

 

$

 

 

$

19,387

 

 

$

142,451

 

 

$

 

 

$

 

 

$

142,451

 

Corporate debt securities

 

 

2,358

 

 

 

 

 

 

 

 

 

2,358

 

Commercial paper

 

 

13,527

 

 

 

 

 

 

(1

)

 

 

13,526

 

 

 

106,879

 

 

 

 

 

 

(115

)

 

 

106,764

 

U.S. Treasury securities

 

 

45,557

 

 

 

9

 

 

 

 

 

 

45,566

 

 

 

3,192

 

 

 

 

 

 

 

 

 

3,192

 

Total cash equivalents

 

 

80,829

 

 

 

9

 

 

 

(1

)

 

 

80,837

 

 

 

252,522

 

 

 

 

 

 

(115

)

 

 

252,407

 

Marketable securities:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

U.S. Treasury securities

 

 

2,991

 

 

 

 

 

 

(7

)

 

 

2,984

 

 

 

23,267

 

 

 

2

 

 

 

 

 

 

23,269

 

Corporate debt securities

 

 

1,034,507

 

 

 

1,143

 

 

 

(9,773

)

 

 

1,025,877

 

 

 

885,237

 

 

 

1,319

 

 

 

(1,885

)

 

 

884,671

 

Certificates of deposit

 

 

81,948

 

 

 

 

 

 

 

 

 

81,948

 

 

 

21,930

 

 

 

 

 

 

 

 

 

21,930

 

Government-sponsored enterprise securities

 

 

145,034

 

 

 

81

 

 

 

(477

)

 

 

144,638

 

 

 

238,595

 

 

 

58

 

 

 

(830

)

 

 

237,823

 

Commercial paper

 

 

289,987

 

 

 

6

 

 

 

(357

)

 

 

289,636

 

 

 

233,124

 

 

 

4

 

 

 

(123

)

 

 

233,005

 

Total marketable securities

 

 

1,554,467

 

 

 

1,230

 

 

 

(10,614

)

 

 

1,545,083

 

 

 

1,402,153

 

 

 

1,383

 

 

 

(2,838

)

 

 

1,400,698

 

Total cash equivalents and marketable securities

 

$

1,635,296

 

 

$

1,239

 

 

$

(10,615

)

 

$

1,625,920

 

 

$

1,654,675

 

 

$

1,383

 

 

$

(2,953

)

 

$

1,653,105

 

December 31, 2022

 

 

 

 

 

 

 

 

 

 

 

 

December 31, 2023

 

 

 

 

 

 

 

 

 

 

 

 

Cash equivalents:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Money market funds

 

$

17,766

 

 

$

 

 

$

 

 

$

17,766

 

 

$

185,990

 

 

$

 

 

$

 

 

$

185,990

 

Corporate debt securities

 

 

2,151

 

 

 

 

 

 

(2

)

 

 

2,149

 

Commercial paper

 

 

32,675

 

 

 

 

 

 

 

 

 

32,675

 

U.S. Treasury securities

 

 

5,731

 

 

 

 

 

 

 

 

 

5,731

 

Total cash equivalents

 

 

52,592

 

 

 

 

 

 

(2

)

 

 

52,590

 

 

 

191,721

 

 

 

 

 

 

 

 

 

191,721

 

Marketable securities:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

U.S. Treasury securities

 

 

22,963

 

 

 

45

 

 

 

 

 

 

23,008

 

Corporate debt securities

 

 

1,236,770

 

 

 

615

 

 

 

(15,006

)

 

 

1,222,379

 

 

 

883,550

 

 

 

3,367

 

 

 

(1,559

)

 

 

885,358

 

Certificates of deposit

 

 

92,417

 

 

 

 

 

 

 

 

 

92,417

 

 

 

47,282

 

 

 

 

 

 

 

 

 

47,282

 

Government-sponsored enterprise securities

 

 

79,746

 

 

 

11

 

 

 

(712

)

 

 

79,045

 

 

 

195,106

 

 

 

377

 

 

 

(352

)

 

 

195,131

 

Commercial paper

 

 

263,231

 

 

 

 

 

 

(509

)

 

 

262,722

 

 

 

155,403

 

 

 

32

 

 

 

(26

)

 

 

155,409

 

Total marketable securities

 

 

1,672,164

 

 

 

626

 

 

 

(16,227

)

 

 

1,656,563

 

 

 

1,304,304

 

 

 

3,821

 

 

 

(1,937

)

 

 

1,306,188

 

Total cash equivalents and marketable securities

 

$

1,724,756

 

 

$

626

 

 

$

(16,229

)

 

$

1,709,153

 

 

$

1,496,025

 

 

$

3,821

 

 

$

(1,937

)

 

$

1,497,909

 

 

As of March 31, 2023 and December 31, 2022,2024 marketable securities were in a net unrealized loss position of $9.41.6 million andmillion. As of December 31, 2023, marketable securities were in a net unrealized gain position of $15.61.9 million, respectively.million. The Company has recorded a net unrealized loss of $3.5 million and a net unrealized gain of $6.2 million and a net unrealized loss of $11.8 million during the three months ended March 31, 20232024 and 2022,2023, respectively, related to its debt securities, which is included in comprehensive loss on the condensed consolidated statements of operations and comprehensive loss.

As of March 31, 20232024 and December 31, 2022,2023, the aggregate fair value of marketable securities that were in an unrealized loss position for less than twelve months was $630.5893.8 million and $628.4463.5 million, respectively. As of March 31, 20232024 and December 31, 2022,2023, the aggregate fair value of marketable securities that were in an unrealized loss position for more than twelve months was $468.0125.9 million and $619.2138.4 million, respectively. Of this amount,As of March 31, 2024, no securities that have been in an unrealized loss position for more than twelve months will mature beyond one year. As of December 31, 2023, securities in an unrealized loss position for more than twelve months totaling $6.32.0 million and $53.1 million as of March 31, 2023 and December 31, 2022, respectively, will maturehad maturities beyond one year.

The Company determined that there is no material credit risk associated with the above investments as of March 31, 2023.2024. The Company has the intent and ability to hold such securities until recovery. As a result, the Company did not record any charges for credit-related impairments for its marketable securities for the three months ended March 31, 2023 and 2022.2024. No available-for-sale debt securities held as of March 31, 20232024 had remaining maturities greater than thirty months.

7


 

3. Fair Value Measurements

The following tables present information about the Company’s financial assets measured at fair value on a recurring basis and indicate the fair value hierarchy classification of such fair values as of March 31, 20232024 and December 31, 20222023 (in thousands):

 

 

Fair Value Measurements at

 

 

Fair Value Measurements at

 

 

March 31, 2023

 

 

March 31, 2024

 

 

Total

 

 

Level 1

 

 

Level 2

 

 

Level 3

 

 

Total

 

 

Level 1

 

 

Level 2

 

 

Level 3

 

Cash and cash equivalents:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Cash

 

$

263,570

 

 

$

263,570

 

 

$

 

 

$

 

 

$

455,020

 

 

$

455,020

 

 

$

 

 

$

 

Money market funds

 

 

19,387

 

 

 

19,387

 

 

 

 

 

 

 

 

 

142,451

 

 

$

142,451

 

 

 

 

 

 

 

Corporate debt securities

 

 

2,358

 

 

 

 

 

 

2,358

 

 

 

 

Commercial paper

 

 

13,526

 

 

 

 

 

 

13,526

 

 

 

 

 

 

106,764

 

 

 

 

 

 

106,764

 

 

 

 

U.S. Treasury securities

 

 

45,566

 

 

 

 

 

 

45,566

 

 

 

 

 

 

3,192

 

 

 

 

 

 

3,192

 

 

 

 

Marketable securities:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

U.S. Treasury securities

 

 

2,984

 

 

 

 

 

 

2,984

 

 

 

 

 

 

23,269

 

 

 

 

 

 

23,269

 

 

 

 

Corporate debt securities

 

 

1,025,877

 

 

 

 

 

 

1,025,877

 

 

 

 

 

 

884,671

 

 

 

 

 

 

884,671

 

 

 

 

Certificates of deposit

 

 

81,948

 

 

 

 

 

 

81,948

 

 

 

 

 

 

21,930

 

 

 

 

 

 

21,930

 

 

 

 

Government-sponsored enterprise securities

 

 

144,638

 

 

 

 

 

 

144,638

 

 

 

 

 

 

237,823

 

 

 

 

 

 

237,823

 

 

 

 

Commercial paper

 

 

289,636

 

 

 

 

 

 

289,636

 

 

 

 

 

 

233,005

 

 

 

 

 

 

233,005

 

 

 

 

Total

 

$

1,889,490

 

 

$

282,957

 

 

$

1,606,533

 

 

$

 

 

$

2,108,125

 

 

$

597,471

 

 

$

1,510,654

 

 

$

 

 

Fair Value Measurements at

 

 

 

 

 

 

 

 

 

 

 

 

 

 

December 31, 2022

 

 

Fair Value Measurements at

 

 

Total

 

 

Level 1

 

 

Level 2

 

 

Level 3

 

 

December 31, 2023

 

 

Total

 

 

Level 1

 

 

Level 2

 

 

Level 3

 

Cash and cash equivalents:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Cash

 

$

159,295

 

 

$

159,295

 

 

$

 

 

$

 

 

$

197,756

 

 

$

197,756

 

 

$

 

 

$

 

Money market funds

 

 

17,766

 

 

 

17,766

 

 

 

 

 

 

 

 

 

185,990

 

 

 

185,990

 

 

 

 

 

 

 

Corporate debt securities

 

 

2,149

 

 

 

 

 

 

2,149

 

 

 

 

Commercial paper

 

 

32,675

 

 

 

 

 

 

32,675

 

 

 

 

U.S. Treasury securities

 

 

5,731

 

 

 

 

 

 

5,731

 

 

 

 

Marketable securities:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

U.S. Treasury securities

 

$

23,008

 

 

 

 

 

 

23,008

 

 

 

 

Corporate debt securities

 

 

1,222,379

 

 

 

 

 

 

1,222,379

 

 

 

 

 

 

885,358

 

 

 

 

 

 

885,358

 

 

 

 

Certificates of deposit

 

 

92,417

 

 

 

 

 

 

92,417

 

 

 

 

 

 

47,282

 

 

 

 

 

 

47,282

 

 

 

 

Government-sponsored enterprise securities

 

 

79,045

 

 

 

 

 

 

79,045

 

 

 

 

 

 

195,131

 

 

 

 

 

 

195,131

 

 

 

 

Commercial paper

 

 

262,722

 

 

 

 

 

 

262,722

 

 

 

 

 

 

155,409

 

 

 

 

 

 

155,409

 

 

 

 

Other non-current assets

 

 

2,212

 

 

 

 

 

 

 

 

 

2,212

 

Total

 

$

1,870,660

 

 

$

177,061

 

 

$

1,691,387

 

 

$

2,212

 

 

$

1,695,665

 

 

$

383,746

 

 

$

1,311,919

 

 

$

 

 

Marketable securities classified as Level 2 within the valuation hierarchy generally consist of U.S. treasury securities and government agency securities, certificates of deposit, corporate bonds and commercial paper. The Company estimates the fair value of these marketable securities by taking into consideration valuations obtained from third-party pricing sources.

4. Property and Equipment, net

Property and equipment, net, consists of the following (in thousands):

 

 

As of

 

 

As of

 

 

March 31,

 

 

December 31,

 

 

March 31,

 

 

December 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Computer equipment

 

$

3,618

 

 

$

3,618

 

 

$

3,739

 

 

$

3,739

 

Furniture, fixtures and other

 

 

8,109

 

 

 

8,109

 

 

 

8,109

 

 

 

8,109

 

Laboratory equipment

 

 

40,176

 

 

 

37,897

 

 

 

41,559

 

 

 

41,411

 

Leasehold improvements

 

 

143,060

 

 

 

141,680

 

 

 

143,260

 

 

 

143,260

 

Construction work in process

 

 

6,104

 

 

 

6,162

 

 

 

8,838

 

 

 

8,859

 

Total property and equipment, gross

 

 

201,067

 

 

 

197,466

 

 

 

205,505

 

 

 

205,378

 

Accumulated depreciation

 

 

(38,869

)

 

 

(33,832

)

 

 

(58,252

)

 

 

(53,433

)

Total property and equipment, net

 

$

162,198

 

 

$

163,634

 

 

$

147,253

 

 

$

151,945

 

 

8


 

Depreciation expense for the three months ended March 31, 2024 and 2023 was $4.8 million and 2022 was $5.0 million, and $respectively.6.0 million, respectively.

5. Accrued Expenses

Accrued expenses consist of the following (in thousands):

 

 

As of

 

 

As of

 

 

March 31,

 

December 31,

 

 

March 31,

 

December 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Payroll and employee-related costs

 

$

8,351

 

 

$

19,241

 

 

$

7,480

 

 

$

17,347

 

Research costs

 

 

22,354

 

 

 

35,010

 

 

 

13,502

 

 

 

16,962

 

Collaboration costs

 

 

44,824

 

 

 

11,177

 

 

 

47,767

 

 

 

2,395

 

Licensing fees

 

 

1,068

 

 

 

983

 

 

 

2,505

 

 

 

3,143

 

Professional fees

 

 

4,041

 

 

 

4,927

 

 

 

3,348

 

 

 

2,515

 

Intellectual property costs

 

 

2,985

 

 

 

3,936

 

 

 

1,363

 

 

 

1,642

 

Accrued property and equipment

 

 

949

 

 

 

1,244

 

 

 

34

 

 

 

630

 

Other

 

 

1,059

 

 

 

1,164

 

 

 

433

 

 

 

701

 

Total

 

$

85,631

 

 

$

77,682

 

 

$

76,432

 

 

$

45,335

 

 

 

6.6. Significant Contracts

Agreements with Vertex

2015 collaboration

In 2015, the Company entered into a strategic collaboration, option and license agreement, or the 2015 Collaboration Agreement, with Vertex Pharmaceuticals Incorporated, or Vertex. The 2015 Collaboration Agreement is focused on the use of the Company’s CRISPR/Cas9 gene editing technology to discover and develop potential new treatments aimed at the underlying genetic causes of human disease. The Company and Vertex amended the 2015 Collaboration Agreement in 2017 and 2019 with Amendment No. 1 and Amendment No. 2, respectively, namely to clarify Vertex’s option rights under the 2015 Collaboration Agreement and to modify certain definitions and provisions of the 2015 Collaboration Agreement to make them consistent with the JDA (as defined below) and the 2019 Collaboration Agreement (as defined below). In 2017, Vertex exercised an option granted to it under the 2015 Collaboration Agreement to obtain a co-exclusive license to develop and commercialize hemoglobinopathy and beta-globin targets, and in 2019, Vertex exercised the remaining options granted to it under the 2015 Collaboration Agreement to exclusively license certain collaboration targets developed under the 2015 Collaboration Agreement.

Exa-celHemoglobinopathies collaboration

In 2017, following Vertex's exercise of its option to obtain a co-exclusive license to develop and commercialize hemoglobinopathy and beta-globin targets, the Company and Vertex entered into a joint development and commercialization agreement, or the JDA, and agreed for potential hemoglobinopathy treatments, including exa-cel,CASGEVY, the Company and Vertex would share equally all research and development costs and worldwide revenues. In 2021, the Company and Vertex amended and restated the JDA, or the A&R Vertex JDCA (as amended and in effect, from time to time), pursuant to which the parties agreed to, among other things, (a) adjust the governance structure for the collaboration and adjust the responsibilities of each party thereunder, whereby Vertex leads and has all decision making (i.e., control) in relation to the exa-celCASGEVY program prospectively; (b) adjust the allocation of net profits and net losses between the parties with respect to exa-celCASGEVY only, which will be allocated 40% to the Company and 60% to Vertex, prospectively; and (c) exclusively license (subject to the Company’s reserved rights to conduct certain activities) certain intellectual property rights to Vertex relating to the specified product candidates and products (including exa-cel)CASGEVY) that may be researched, developed, manufactured and commercialized on a worldwide basis under the A&R Vertex JDCA. Additionally, the A&R Vertex JDCA allows the Company to defer a portion of its share of costs under the arrangement if spending on the exa-celCASGEVY program exceeds specified amounts.amounts through 2024. In December 2023, the Company entered into an amendment to the A&R Vertex JDCA, or Amendment No. 1 to the A&R Vertex JDCA, with Vertex related to the global development, manufacturing, and commercialization of CASGEVY. Pursuant to Amendment No. 1 to the A&R Vertex JDCA, among other things, the Company and Vertex agreed to (a) allocate certain costs arising from a license agreement with a third party, resulting in a current payment due to Vertex by the Company of $20.0 million upon an event specified in Amendment No. 1 to the A&R Vertex JDCA, and (b) adjust, under certain specified circumstances, the timing of and portion of the Company’s share of costs it is permitted to defer under the agreement. Any deferred amounts under the A&R Vertex JDCA, as amended, are only payable to Vertex as an offset against future profitability of the exa-celCASGEVY program and the amounts payable are capped at a specified maximum amount per year.

9


In connection with the closing of the transaction contemplated by the A&R Vertex JDCA, the Company received a $900.0 million up-front payment from Vertex. Additionally, in December 2023, the Company and Vertex received approval of CASGEVY by the U.S. Food and Drug Administration, or the FDA. The FDA’s approval of CASGEVY triggered Vertex’s obligation to make a $200.0 million milestone payment to the Company, which was recognized in December 2023 and for which payment was received in January 2024 is included in accounts receivable in the accompanying consolidated balance sheets as of December 31, 2023.

DMD and DM1 exclusive license

In 2019, the Company and Vertex entered into a series of agreements, including a strategic collaboration and license agreement, or the 2019 Collaboration Agreement, for the development and commercialization of products for the treatment of Duchenne muscular dystrophy, or DMD, and Myotonic Dystrophymyotonic dystrophy Type 1, or DM1. For the DMD and DM1 programs, Vertex is responsible for all research, development, manufacturing and commercialization activities and all related costs. Upon IND filing, the Company has the option to forego the DM1 milestones and royalties, and instead, co-develop and co-commercialize all DM1 products globally in

9


exchange for payment of 50% of research and development costs incurred by Vertex from the effective date of the agreement through IND filing.

Collaboration in the field of diabetes

In 2021, CRISPRthe Company and ViaCyte, Inc., or ViaCyte, entered into a joint development and commercialization agreement, or the ViaCyte JDCA, to jointly develop and commercialize product candidates and shared products for the diagnosis, treatment or prevention of diabetes type 1, diabetes type 2 or insulin dependent / requiring diabetes throughout the world. In the third quarter of 2022, Vertex acquired ViaCyte, and ViaCyte became a wholly-owned subsidiary of Vertex. In March 2023, (1) the Company and ViaCyte entered into an amendment to the ViaCyte JDCA, or the ViaCyte JDCA Amendment, and adjusted certain rights and obligations of the Company and ViaCyte under the ViaCyte JDCA, and (2) the Company and Vertex entered into a non-exclusive license agreement, or the Non-Ex License Agreement, pursuant to which the Company agreed to license to Vertex, on a non-exclusive basis, certain of its gene editing intellectual property to exploit certain products for the diagnosis, treatment or prevention of diabetes type 1, diabetes type 2 or insulin dependent / requiring diabetes throughout the world. Subsequently, ViaCyte elected to opt-out of the ViaCyte JDCA. Per the opt-out terms, the on-going collaboration assets are now wholly owned by the Company, subject to a royalty on future sales owed to ViaCyte. The opt-out became effective in early February 2024.

In connection with entering into these agreements,the Non-Ex License Agreement, the Company received a $100.0 million up front payment from Vertex. UnderVertex in the Non-Ex License Agreement,first quarter of 2023 and subsequently received a $70.0 million research milestone achieved in the second quarter of 2023. The Company is eligible to receive additional milestone payments from Vertex of up to $230.0160 million in the aggregate, dependingwhich are dependent on the achievement of pre-determined research, development and commercial milestones for certain products utilizing the licensed intellectual property. Additionally, the Company is eligible to receive tiered royalties on the sales of certain products in the low to mid-single digits.

Accounting Analysis

TheFor purposes of this Note 6, the 2015 Collaboration Agreement, Amendment No. 1, Amendment No. 2, A&R Vertex JDCA, Amendment No. 1 to the A&R Vertex JDCA and 2019 Collaboration Agreement are collectively referred to as the “Vertex Agreements” for purposes of this Note 6. Theand the Non-Ex License Agreement and ViaCyte JDCA Amendment are collectively referred to as the “March 2023 Agreements.”

The Vertex Agreements and the March 2023 Agreements include components of a customer-vendor relationship as defined under ASC 606, Revenue from Contracts with Customers, or ASC 606, collaborative arrangements as defined under ASC 808, Collaborative Agreements, or ASC 808, and research and development costs as defined under ASC 730, Research and Development, or ASC 730. Specifically, with regards to the March 2023 Agreements, the Company concluded that the non-exclusive license is a performance obligation under ASC 606 and the ongoing research and development services under the ViaCyte JDCA Amendment are a unit of account under ASC 808.

Accounting Analysis Under ASC 606

March 2023 Agreements

Identification of the contractContract

The March 2023 Agreements were negotiated as a package with a single commercial objective and, as such, the March 2023 Agreements were combined for accounting purposes and treated as a single arrangement. The Company determined for accounting purposes that the combined contract terminates the original ViaCyte JDCA and created a new contract.

10


Identification of performance obligationsPerformance Obligations

The Company concluded the transfer of the non-exclusive license, including certain modified rights and obligations provided as part of the ViaCyte JDCA Amendment to support the delivery of the license, was both capable of being distinct and distinct within the context of the contract.

Determination of transaction priceTransaction Price

The initial transaction price was comprised of the upfront payment of $100.0 million.

In the second quarter of 2023, the Company adjusted the transaction price to include $70.0 million in previously constrained variable consideration related to a research milestone which was achieved in the second quarter of 2023. The Company determined that all other possible variable consideration resulting from milestones and royalties discussed below was fully constrained at the timeas of the transaction.March 31, 2024. The Company will reevaluatere-evaluate the transaction price in each reporting period.

10


Allocation of transaction priceTransaction Price to performance obligationsPerformance Obligations

The Company identified one performance obligation for the March 2023 Agreements and, as a result, no allocation of the transaction price was required.

Recognition of revenueRevenue

The Company determined the non-exclusive license, including certain modified rights and obligations provided as part of the ViaCyte JDCA Amendment to support the delivery of the license, represented functional intellectual property, as the intellectual property provides Vertex with the ability to perform a function or task in the form of research and development in the field of diabetes. TheIn 2023, the Company recognized revenue of $100.0 million for the non-exclusive license at the onset of the arrangement, as this was the point in time in which the non-exclusive license was delivered.

In 2023, revenue from variable consideration of $70.0 million was recognized related to a research milestone that was achieved during the second quarter of 2023. Revenue recognized under the March 2023 Agreements for year ended December 31, 2023 was $100.0170.0 million in aggregate.

No revenue was recognized under the March 2023 Agreements for the three months ended March 31, 2023.2024. Revenue recognized under the March 2023 Agreements for the three months ended March 31, 2023 was $100.0 million.

Milestones under the Non-Ex License Agreement

TheAs of March 31, 2024, the Company is eligible to receive potential future milestone payments from Vertex of up to $230.0160.0 million in the aggregate under the Non-Ex License Agreement depending on the achievement of pre-determined research, development and commercial milestones for certain products utilizing the licensed intellectual property. Additionally, the Company is eligible to receive tiered royalties on the sales of certain products in the low to mid-single digits.

Each of the remaining milestones under the Non-Ex License Agreement are fully constrained as of March 31, 2023.2024. There is uncertainty as to whether the events to obtain the research and developmental milestones will be achieved given the nature of clinical development and the stage of the CRISPR/Cas9 technology. The remaining research, development and regulatory milestones will be constrained until it is probable that a significant revenue reversal will not occur. Commercial milestones and royalties relate predominantly to a license of intellectual property and are determined by sales or usage-based thresholds. The commercial milestones and royalties are accounted for under the royalty recognition constraint and will be accounted for as constrained variable consideration. The Company applies the royalty recognition constraint for each commercial milestone and will not recognize revenue for each until the subsequent sale of a licensed product (achievement of each) occurs.

Vertex Agreements

Deferred revenue

As of March 31, 20232024 and December 31, 2022,2023, there was no current deferred revenue related to the Vertex Agreements. As of March 31, 2023,2024, there was $12.3 million of non-current deferred revenue related to the Vertex Agreements, which is unchanged from December 31, 2022.2023. The transaction price allocated to the remaining performance obligations was $12.3 million.

Milestones

The Company has evaluated the milestones that may be received in connection with the Vertex Agreements.

Under the 2015 Collaboration Agreement and subsequent amendments, the Company is eligible to receive up to $410.0 million in additional development, regulatory and commercial milestones and royalties on net product sales for each of the three collaboration targets that Vertex licensed in 2019. Each milestone is payable only once per collaboration target, regardless of the number of products directed to such collaboration target that achieve the relevant milestone event.

11


The Company is eligible to receive potential future payments of up to $775.0 million under the 2019 Collaboration Agreement based upon the successful achievement of specified development, regulatory and commercial milestones for the DMD and DM1 programs. The Company is also eligible to receive tiered royalties on future net sales on any products that may result from this collaboration; however, the Company has the option to forego the DM1 milestones and royalties to co-develop and co-commercialize all DM1 products globally.

The Company is eligible to receive potential future payments of up to $200.0 million under the A&R Vertex JDCA upon receipt by Vertex of the first marketing approval of the initial product candidate from the FDA or the European Commission. In addition, the Company has the option to conduct research at their own cost in certain defined areas that, if beneficial to the exa-celCASGEVY program and exa-celCASGEVY ultimately achieves regulatory approval in such areas, then the Company could be entitled to certain milestone payments aggregating to high eight digits from Vertex.

Each of the remaining milestones described above are fully constrained as of March 31, 2023.2024. There is uncertainty that the events to obtain the research and developmental milestones will be achieved given the nature of clinical development and the stage of the CRISPR/Cas9 technology. The remaining research, development and regulatory milestones will be constrained until it is probable that a significant revenue reversal will not occur. Commercial milestones and royalties relate predominantly to a license of intellectual property and are determined by sales or usage-based thresholds. The commercial milestones and royalties are accounted for under the royalty recognition constraint and will be accounted for as constrained variable consideration. The Company applies the royalty recognition constraint for each commercial milestone and will not recognize revenue for each until the subsequent sale of a licensed product (achievement of each) occurs.

11


Accounting Analysis under ASC 808

Vertex Agreements

In connection with the Vertex Agreements, the Company identified the following collaborative elements, which are accounted for under ASC 808: (i) development and commercialization services for shared products, including any transition services related to exa-celCASGEVY under the A&R Vertex JDCA; (ii) R&D Services for follow-on products; and (iii) committee participation. The related impact of the cost sharing is included within collaboration expense, net, in the condensed consolidated statements of operations and comprehensive loss. During the three months ended March 31, 20232024 and 2022,2023, the Company recognized $42.247.0 million and $30.642.2 million of collaboration expense, net, related to the exa-celCASGEVY program, respectively. Collaboration expense, net, during the three months ended March 31, 2024 and 2023 was net of $2.80.8 million and $7.42.8 million of reimbursements from Vertex related to the exa-celCASGEVY program, respectively.

 

12


 

7.7. Commitments and Contingencies

Leases

Refer to Note 7 to the consolidated financial statements in the Company’s 20222023 Annual Report on Form 10-K filed with the SEC on February 21, 20232024 for discussion of the Company’s lease arrangements.

Litigation

In the ordinary course of business, the Company is from time to time involved in lawsuits, investigations, proceedings and threats of litigation related to, among other things, the Company’s intellectual property estate (including certain in-licensed intellectual property), commercial arrangements and other matters. Such proceedings may include quasi-litigation, inter partes administrative proceedings in the U.S. Patent and Trademark Office and the European Patent Office involving the Company’s intellectual property estate including certain in-licensed intellectual property. The outcome of any of the foregoing, regardless of the merits, is inherently uncertain. In addition, litigation and related matters are costly and may divert the attention of Company’s management and other resources that would otherwise be engaged in other activities. If the Company is unable to prevail in any such proceedings, the Company’s business, results of operations, liquidity and financial condition could be adversely affected.

Letters of Credit

As of March 31, 2023,2024, the Company had restricted cash of $11.711.5 million, representing letters of credit securing the Company’s obligations under certain leased facilities. The letters of credit are secured by cash held in a restricted depository account and included in “Restricted cash” on the Company's condensed consolidated balance sheets as of March 31, 2023.2024.

Research, Manufacturing, License and Intellectual Property Agreements

The Company has engaged several research institutions and companies to identify new delivery strategies and applications of the Company’s gene editing technology. The Company is also a party to a number of license agreements which require significant upfront payments and may be required to make future royalty payments and potential milestone payments from time to time. In addition, the Company is also a party to intellectual property agreements, which require maintenance and milestone payments from time to time. Further, the Company is a party to a number of manufacturing agreements that require upfront payments for the future performance of services.

In association with these agreements, on a product-by-product basis, the counterparties are eligible to receive up to low eight-digit potential payments upon specified research, development and regulatory milestones. In addition, on a product-by-product basis, the counterparties are eligible to receive potential commercial milestone payments based on specified annual sales thresholds. The potential payments are low-single digit percentages of the specified annual sales thresholds. The counterparties are also eligible to receive low single-digit royalties on future net sales.

Under certain circumstances and if certain contingent future events occur, Vertex is eligible to receive up to $395.0 million in potential specified research, development, regulatory and commercial milestones and tiered single-digit percentage royalties on future net sales related to a specified target under an amendment to the 2015 Collaboration Agreement (as such term is defined in Note 6 above). In addition, Vertex has the option to conduct research at its own cost in certain defined areas that, if beneficial to the exa-celCASGEVY program and ultimately achieves regulatory approval, could result in the Company owing Vertex certain milestone payments aggregating to high eight digits, subject to certain limitations on the profitability of the exa-celCASGEVY program.

12


Under the A&R Vertex JDCA, the Company deferredhas an option to defer specified costs on the CASGEVY program in excess of $110.3 million for the years ended December 31, 2022, 2023 and 2024. As of March 31, 2024, $4.0 million of costs incurred in 2024 under the A&R JDCA have been deferred. In 2023, the Company exercised its option to defer specified costs on the CASGEVY program in excess of the deferral limit under A&R Vertex JDCA, which resulted in a deferral of $80.9 million as of December 31, 2023. In 2022, the Company also exercised its option to defer its share of costs incurred in 2022 on the CASGEVY program in excess of the deferral limit under the A&R Vertex JDCA, which resulted in a deferral of $36.1 million as of its share of costs incurred under the arrangement for the year ended December 31, 2022, as spending on the exa-cel program exceeded a specified amount.2022. Any deferred amounts are only payable to Vertex as an offset against future profitability of the exa-celCASGEVY program and the amounts payable are capped at a specified maximum amount per year. These deferred costs on the exa-celCASGEVY program will be accrued for when it is probable that a liability has been incurred and the amount can be reasonably estimated. As of March 31, 2023,2024, no contingent payments have been accrued. Referaccrued to Note 6 for further discussion of the Company’s arrangements with Vertex.date.

13


8. Share Capital

The Company had 150,347,467 authorizedAll of the Company's common shares are authorized under Swiss corporate law with a nominal value of 0.03 CHF per share. Though the nominal value of common shares is stated in Swiss francs, the Company continues to use U.S. dollars as its reporting currency for preparing the condensed consolidated financial statements.

As of March 31, 2023,2024, the Company's share capital consists of 88,517,810 registered common shares with a parnominal value of CHF 0.03 per share. Share Capital consistedshare, 8,202,832 registered common shares reserved for potential issuance of bonds or similar instruments, and 19,925,932 registered common shares reserved for the Company's employee equity incentive plans. In addition, the Board of Directors is authorized to conduct one or more increases of the following:share capital at any time until June 8, 2028, or the expiry of the capital band if earlier, within a lower limit of CHF 2,538,051.84 and an upper limit of CHF 2,952,222.57, corresponding to 13,805,691 registered common shares with a nominal value of CHF 0.03 each to be fully paid in.

 

 

 

 

 

As of

 

Type of Share Capital

 

Conditional Capital

 

March 31, 2023

 

 

December 31, 2022

 

Common shares

 

Registered share capital

 

 

83,538,347

 

 

 

82,028,328

 

Common shares

 

Authorized share capital

 

 

39,316,975

 

 

 

39,316,975

 

Common shares

 

Conditional share capital - Bonds or similar debt instruments

 

 

8,202,832

 

 

 

8,202,832

 

Common shares

 

Conditional share capital - Employee benefit plans

 

 

19,289,313

 

 

 

20,799,332

 

 

 

Total

 

 

150,347,467

 

 

 

150,347,467

 

Common Share Issuances

At-the-Market Offering

In August 2019, the Company entered into an Open Market Sale AgreementSM with Jefferies LLC, or Jefferies, under which the Company was able to offer and sell, from time to time at its sole discretion through Jefferies, as its sales agent, its common shares, or the August 2019 Sales Agreement.

In January 2021, in connection with the August 2019 Sales Agreement, the Company filed a prospectus supplement with the SEC to offer and sell, from time to time, common shares having aggregate gross proceeds of up to $600.0 million. In July 2021, the Company filed a new prospectus supplement with the SEC, which replaced the previous prospectus supplement filed in January 2021, to offer and sell, from time to time, the common shares remaining under the original prospectus supplement having aggregate gross proceeds of up to $419.8 million, or, together with the January 2021 prospectus supplement, the 2021 ATM.

As of March 31, 2023,2024, the Company has issued and sold an aggregate of 1.11.5 million common shares under the 2021 ATM at an average price of $168.79139.28 per share for aggregate proceeds of $178.8212.4 million, which were net of equity issuance costs of $2.42.9 million, excluding stamp taxes.

Registered Direct Offering

In February 2024, the Company entered into an investment agreement for the sale of approximately $280.0 million of its common shares to a group of institutional investors in a registered direct offering, at a price per share of $71.50. The Company received net proceeds of $279.0 million, excluding stamp taxes due of $2.8 million.

9. Stock-based Compensation

During the three months ended March 31, 20232024 and 2022,2023, the Company recognized the following stock-based compensation expense (in thousands):

 

 

Three Months Ended March 31,

 

 

Three Months Ended March 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Research and development

 

$

11,676

 

 

$

14,589

 

 

$

11,182

 

 

$

11,676

 

General and administrative

 

 

9,199

 

 

 

11,156

 

 

 

8,223

 

 

 

9,199

 

Total

 

$

20,875

 

 

$

25,745

 

 

$

19,405

 

 

$

20,875

 

 

1314


 

 

Stock option activity

The following table summarizes stock option activity for the three months ended March 31, 2023:2024:

 

 

 

Shares

 

 

Weighted-
average
exercise price
per share

 

Outstanding at December 31, 2022

 

 

7,230,233

 

 

$

60.22

 

Granted

 

 

1,236,882

 

 

 

44.05

 

Exercised

 

 

(159,184

)

 

 

30.37

 

Cancelled or forfeited

 

 

(390,947

)

 

 

86.12

 

Outstanding at March 31, 2023

 

 

7,916,984

 

 

$

57.01

 

Exercisable at March 31, 2023

 

 

4,853,602

 

 

$

52.83

 

Vested and expected to vest at March 31, 2023

 

 

7,916,984

 

 

$

57.01

 

 

 

Shares

 

 

Weighted-
average
exercise price
per share

 

Outstanding at December 31, 2023

 

 

7,204,372

 

 

$

55.05

 

Granted

 

 

811,624

 

 

 

72.94

 

Exercised

 

 

(632,683

)

 

 

38.72

 

Cancelled or forfeited

 

 

(218,964

)

 

 

71.20

 

Outstanding at March 31, 2024

 

 

7,164,349

 

 

$

58.03

 

Exercisable at March 31, 2024

 

 

4,471,078

 

 

$

55.62

 

Vested and expected to vest at March 31, 2024

 

 

7,164,349

 

 

$

58.03

 

 

 

As of March 31, 2023,2024, total unrecognized compensation expense related to stock options was $117.4101.7 million, which the Company expects to recognize over a remaining weighted-average period of 2.62.8 years.

 

Restricted stock activity

The following table summarizes restricted stock activity for the three months ended March 31, 2023:2024:

 

 

Shares

 

 

Weighted-
Average
Grant Date
Fair Value

 

 

Shares

 

 

Weighted-
Average
Grant Date
Fair Value

 

Unvested balance at December 31, 2022

 

 

1,325,185

 

 

$

80.13

 

Unvested balance at December 31, 2023

 

 

1,781,415

 

 

$

61.00

 

Granted

 

 

643,724

 

 

 

43.76

 

 

 

603,994

 

 

 

72.94

 

Vested

 

 

(172,995

)

 

 

74.21

 

 

 

(214,913

)

 

 

61.19

 

Cancelled or forfeited

 

 

(125,084

)

 

 

86.11

 

 

 

(88,464

)

 

 

53.19

 

Unvested balance at March 31, 2023

 

 

1,670,830

 

 

$

66.29

 

Unvested balance at March 31, 2024

 

 

2,082,032

 

 

$

64.77

 

 

As of March 31, 2023,2024, total unrecognized compensation expense related to unvested restricted common shares was $82.0103.2 million, which the Company expects to recognize over a remaining weighted-average vesting period of 2.83.0 years.

10. Net Loss Per Share Attributable to Common Shareholders

Basic net loss per share is calculated by dividing net loss attributable to common shareholders by the weighted-average number of common shares outstanding during the period. Diluted net loss per share is calculated by dividing the net loss attributable to common shareholders by the weighted-average number of common share equivalents outstanding for the period, including any dilutive effect from outstanding stock options and warrants using the treasury stock method. The Company’s net loss is net loss attributable to common shareholders for all periods presented.

The following common stock equivalents were excluded from the calculation of diluted net loss per share for the periods indicated because including them would have had an anti-dilutive effect (in thousands):effect:

 

 

Three Months Ended March 31,

 

 

Three Months Ended March 31,

 

 

2023

 

 

2022

 

 

2024

 

 

2023

 

Outstanding options

 

 

7,916,984

 

 

 

8,220,458

 

 

 

7,164,349

 

 

 

7,916,984

 

Unvested restricted common shares

 

 

1,670,830

 

 

 

1,147,364

 

 

 

2,082,032

 

 

 

1,670,830

 

ESPP

 

 

18,110

 

 

 

10,969

 

 

 

8,630

 

 

 

18,110

 

Total

 

 

9,605,924

 

 

 

9,378,791

 

 

 

9,255,011

 

 

 

9,605,924

 

 

 

 

1415


 

11. Income Taxes

During the three months ended March 31, 20232024 and 2022,2023, the Company recorded an income tax provision of $1.30.7 million and $3.61.3 million, respectively, representing an effective tax rate of (2.60.6%) and (2.12.6%), respectively. The income tax provision for the three months ended March 31, 2023 and 20222024 is primarily attributable to the income generated by the Company's U.S. subsidiaries. The change in the rate for the three months ended March 31, 20232024 is primarily attributable to reduced forecasted capitalized R&D expense addback offset by an increase in forecasted interest income in the United States. The difference in the statutory tax rate and effective tax rate is primarily a result of the jurisdictional mix of earnings, research credits generated, and the valuation allowance recorded against certain deferred tax assets. The Company maintains a valuation allowance against certain deferred tax assets that are not more-likely-than-not realizable.realizable

1516


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

The following discussion and analysis of our financial condition and results of operations should be read in conjunction with (i) our unaudited condensed consolidated financial statements and related notes appearing elsewhere in this Quarterly Report on Form 10-Q and (ii) our audited consolidated financial statements and related notes and management’s discussion and analysis of financial condition and results of operations included in our Annual Report on Form 10-K for the year ended December 31, 20222023 filed with the Securities and Exchange Commission, or the SEC, on February 21, 2023.2024. 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 and impact and potential impacts on our business, includes forward-looking statements that involve risks and uncertainties. As a result of many factors, including, without limitation, those factors set forth in the “Risk Factors” section of our Annual Report on Form 10-K for the year ended December 31, 20222023 and the “Risk Factors” section of subsequent Quarterly Reports on Form 10-Q, our actual results or timing of certain events could differ materially from the results or timing described in, or implied by, these forward-looking statements.

Overview

We are a leading gene editing company focused on the development of CRISPR/Cas9-based therapeutics. CRISPR/Cas9 is a revolutionary gene editing technology that allows for precise, directed changes to genomic DNA. The application of CRISPR/Cas9 for gene editing, was co-invented by onethe process of our scientific founders, Dr. Emmanuelle Charpentier. Dr. Charpentier and her collaborators published work elucidating how CRISPR/Cas9, a naturally occurring viral defense mechanism found in bacteria, can be adapted for use in gene editing.precisely altering specific sequences of genomic DNA. We are applyingaim to apply this technology to disrupt, delete, correct and insert genes to potentially treat genetically defined diseases and to engineer advanced cellular therapies. We have advanced this technology from discovery to an approved medicine with unparalleled speed, culminating in the landmark first approval of a CRISPR-based therapy, CASGEVY (exagamglogene autotemcel [exa-cel]), in 2023 with our collaborators at Vertex Pharmaceuticals Incorporated, or Vertex. We believe that the combination of our scientific expertise, together with our gene editing approach,technology, research and development capabilities, and proven ability to execute may enable us to create an entirely new class of highly effective and potentially curative therapies for patients with both rare and common diseases for whom current biopharmaceutical approaches have had limited success.

We have established a portfolio of therapeutic programs in a broad range of disease areas acrossspanning four core franchises: hemoglobinopathies, immuno-oncology, regenerative medicine and autoimmune iinn vivo approaches. approaches and Type 1 diabetes. Our most advanced programs targetprogram, CASGEVY, has received approval in the genetically defined diseases transfusion-dependent beta thalassemia, or TDT,United States and other countries for the treatment of eligible patients with severe sickle cell disease, or SCD, or transfusion-dependent beta thalassemia, or TDT, two genetic disorders of hemoglobin, or hemoglobinopathies, with high unmet medical need. In addition, we have further research efforts on targeted conditioning and in vivo editing of hematopoietic stem cells that have the potential to expand the number of patients that could benefit significantly.We are also progressing severalmultiple next-generation gene-edited allogeneic cell therapy programs, including allogeneic chimeric antigen receptor T cell, or CAR T, candidates for the treatment of hematological and solid tumor cancers and investigational, allogeneic, gene-edited, immune-evasive, stem cell-derived therapies for the treatment of type 1 diabetes, or T1D.autoimmune diseases. In addition, we are advancing multiplea portfolio of programs leveraging in vivoediting approaches, initially for both common and rare diseases, starting with the treatment and prevention of cardiovascular disease. Further, we have multiple parallel efforts using allogeneic, gene-edited, hypoimmune, stem cell-derived beta cells to address type 1 diabetes, or T1D, without the need for chronic immunosuppression.

Hemoglobinopathies

Our lead product candidate, exa-cel,CASGEVY

CASGEVY is an investigational, autologous,a non-viral, ex vivo CRISPRCRISPR/Cas9 gene-edited cell therapy, in which a patient’s own hematopoietic stem celland progenitor cells are edited at the erythroid specific enhancer region of the BCL11A gene through a precise double-strand break. This edit results in the production of high levels of fetal hemoglobin in red blood cells, which can compensate for the defective adult hemoglobin in patients with SCD and TDT. CASGEVY is the first therapy thatto emerge from our strategic partnership with Vertex and is being evaluated for the treatment of TDT and severe SCD. Exa-cel is being developedadvanced under a joint development and commercialization agreement between us and Vertex. WeVertex and Vertex are investigating exa-cel in two ongoing Phase 1/2/3 open-label clinical trials that are designed to assess the safety and efficacycertain of a single dose of exa-cel in patients ages 12 to 35 with TDT (CLIMB-111) or severe SCD (CLIMB-121), respectively. Enrollment is complete for both CLIMB-111 and CLIMB-121. In the second and fourth quarters of 2022, at the European Hematology Association Congress and American Society of Hematology Annual Meeting, respectively, we presented updated clinical data from CLIMB-111 and CLIMB-121 for 44 patients with TDT and 31 patients with severe SCD treated with exa-cel. In addition, we and Vertex are conducting two additional Phase 3 open-label clinical trials of exa-cel in pediatric patients with TDT (CLIMB-141) and severe SCD (CLIMB-151). Patients who received exa-cel in CLIMB-111, CLIMB-121, CLIMB-141 or CLIMB-151 are followed for approximately two years after exa-cel infusion and are asked to participate in a long-term, open-label follow-up trial, CLIMB-131, to evaluate the safety and efficacy of exa-cel. CLIMB-131 is designed to follow participants for up to 15 years after exa-cel infusion.its affiliates.

We and Vertex recently completed rolling submissions of

In 2023, CASGEVY became the Biologics Licensing Applications, or BLAs, for exa-celfirst-ever approved CRISPR-based gene-editing therapy in the world. To date, CASGEVY has been approved in the United States. Exa-cel has been granted RMAT, Fast Track, Orphan Drug,States, Europe, Great Britain, Saudi Arabia and Rare Pediatric Disease designations by the FDABahrain for the treatment of both TDTeligible patients 12 years and SCD. In addition, inolder with SCD or TDT. Efficacy data presented to date support the fourth quarterprofile of 2022, wethis therapy as a potential one-time functional cure for people with severe SCD and Vertex completed regulatory submissions for exa-cel in the EU and the United Kingdom, and the European Medicines Agency, or EMA, and the Medicines and Healthcare products Regulatory Agency, or MHRA, have validated the marketing applications, respectively, indicating acceptance of the marketing applications and initiation of the review. Exa-cel has also been granted Orphan Drug Designation from the European Commission, as well as PRIME designation from the EMA for the treatment of both TDT and SCD. In the United Kingdom, exa-cel has been granted an Innovation Passport under the Innovative Licensing and Access Pathway from the MHRA.TDT.

In addition, buildingAdditional candidates

Building upon exa-cel,CASGEVY, we have next-generation efforts in targeted conditioning and in vivo editing of hematopoietic stem cells, either of which could broaden the number of patients that cancould benefit from our therapies.hemoglobinopathies product candidates.

1617


 

Immuno-Oncology and Autoimmune

We believe CRISPR/Cas9 has the potential to create the next generation of CAR T cell therapies that may have a superior product profile and allow broader patient access compared to current autologous therapies and allow accessibility to broader patient populations. Drawing from thetherapies. ex vivo gene editing capabilities gained through our lead programs, weWe are advancing several immuno-oncology cell therapy programs for oncology and/or autoimmune indications, including two next-generation allogeneic CAR T programs, CTX112 targeting Cluster of Differentiation 19, or CD19 and ClusterCTX131 targeting CD70. These product candidates incorporate two novel gene edits—knock-out of Differentiation 70,Regnase-1 and knock-out of transforming growth factor-beta receptor type 2, or CD70.TGFBR2—designed to enhance CAR T potency and reduce CAR T exhaustion. Emerging pharmacology data, including pharmacokinetics, from ongoing clinical trials of CTX112 and CTX131, indicate that the novel potency gene edits lead to significantly higher CAR T cell expansion and functional persistence in patients compared to our first-generation candidates that did not incorporate these edits. In addition, the next-generation candidates exhibit increased manufacturing robustness, with a higher and more consistent number of CAR T cells produced per batch. We are producing CTX112 and CTX131 for clinical trials at our internal GMP manufacturing facility.

CD19 FranchiseCandidates

CTX110, our lead immuno-oncology product candidate,

CTX112 is a healthy donor-derived gene-edited allogeneic CAR T investigational therapy targeting CD19. We are investigating CTX110being investigated in our CARBONan ongoing clinical trial which is designed to assess the safety and efficacy of CTX110the candidate in adult patients with relapsed or refractory CD19-positive B-cell malignancies who have received at least two prior lines of therapy. CTX110 has been granted RMAT designation by the FDA.

The Phase 1 CARBONIn addition, we plan to initiate a clinical trial is being conductedof CTX112 in two parts – Part A and Part B. In Part Asystemic lupus erythematosus in the first half of the Phase 1 CARBON clinical trial, or Phase 1 Part A, patients were infused with a single dose of CTX110 across escalating dose levels following a standard lymphodepletion regimen, with an option to re-dose CTX110 based on clinical benefit. In Part B of the Phase 1 CARBON clinical trial, or Phase 1 Part B, patients received CTX110 at Dose Level 4 following standard lymphodepletion, as well as a consolidation dose of CTX110 at the same dose level between four and eight weeks after the initial dose for patients that demonstrated clinical benefit.

In the fourth quarter of 2022, we presented updated clinical data from Phase 1 Part A for 32 patients treated with CTX110, which showed the potential for CTX110 to achieve long-term durable complete remissions with a positively differentiated safety profile in heavily pre-treated patients, and described emerging data from Phase 1 Part B, which showed an encouraging efficacy profile2024, with the potential to improve efficacy withexpand into additional autoimmune indications in the usefuture. Early clinical studies conducted by third parties have shown that CD19-directed autologous CAR T therapy can produce long-lasting remissions in multiple autoimmune indications by deeply depleting B cells. Our first generation allogeneic CD19-directed CAR T program has demonstrated effective depletion of a consolidation dose. Based on this emerging data from our Phase 1 CARBON clinical trial and discussions with regulatory agencies, we have expanded CARBON to include a Phase 2, potentially registrational, single-arm, multi-center, open-label clinical trial that incorporates consolidation dosing. The Phase 2 clinical trial is ongoing.B cells in oncology settings, which supports the potential for CTX112 in autoimmune diseases.

In parallel with CTX110, we are advancing CTX112, a next-generation investigational, allogeneic CAR T product candidate targeting CD19. CTX112 incorporates additional edits designed to enhance CAR T potency and reduce CAR T exhaustion. In the fourth quarter of 2022, the IND for CTX112 was cleared by the FDA. CTX112CD70 Candidates

CTX131 is being investigated in an ongoing Phase 1/2 clinical trial designed to assess the safety and efficacy of CTX112 in adult patients with relapsed or refractory CD19-positive B-cell malignancies who have received at least two prior lines of therapy.

CD70 Franchise

CTX130 is a healthy donor-derived gene-edited allogeneic CAR T investigational therapy targeting CD70, an antigen expressed on various solid tumors and hematologic malignancies. CTX130 is being investigated in two ongoing independent Phase 1, single-arm, multi-center, open-label clinical trials, COBALT, that are designed to assess the safety and efficacy of several dose levels of CTX130 in adult patients. The COBALT-LYM trial is evaluating the safety and efficacy of CTX130 for the treatment of relapsed or refractory T or B cell malignancies. The COBALT-RCC trial is evaluating the safety and efficacy of CTX130 for the treatment of relapsed or refractory clear cell renal cell carcinoma. CTX130 has received Orphan Drug Designation from the FDA for the treatment of T cell lymphoma and RMAT designation for the treatment of Mycosis Fungoides and Sézary Syndrome (MF/SS), subtypes of Cutaneous T cell Lymphoma (CTCL). In the second quarter of 2022, at the European Hematology Association Congress, we released initial clinical data from the ongoing COBALT-LYM trial for 18 patients with T cell lymphoma treated with CTX130 who had reached at least 28 days of follow-up. Also, in the fourth quarter of 2022, at the Society of Immuno-therapy in Cancer Annual Meeting, we released initial clinical data from the COBALT-RCC trial for 14 patients.

In parallel with CTX130, we are advancing CTX131, a next-generation investigational allogeneic CAR T product candidate targeting CD70 in a basket of solid tumors. CTX131 incorporates additional edits designed to enhance CAR T potency and reduce CAR T exhaustion. In the first quarter of 2023, the IND for CTX131 was cleared by the FDA. CTX131 is being investigated in an ongoing Phase 1/2 clinical trial designed to assess the safety and efficacy of CTX131 in adult patients with relapsed or refractory solid tumors. In addition, we plan to initiate a clinical trial of CTX131 in hematologic malignancies in the first half of 2024.

Additional candidates

Our CRISPR/Cas9 platform enables us to innovate continuously by incorporating incremental edits into next-generation products. In addition to CTX112 and CTX131, weWe are advancing several additional investigational CAR T product candidates.

Regenerative Medicine

Regenerative medicine, or the use of stem cells to repair or replace tissue or organ function lost due to disease, damage or age, holds the potential to treat both rare and common diseases. Building upon our ex vivo gene editing expertise, we have expanded our efforts in this field with a focus on allogeneic stem cell-derived therapies gene edited using CRISPR/Cas9 to enable immune evasion,

17


improve cell function, and direct cell fate. Our first major effort in this area is in diabetes, and we and ViaCyte, Inc., or ViaCyte, which was acquired by Vertex in the third quarter of 2022, are advancing a series of programs as part of a strategic collaboration for the discovery, development, and commercialization of gene-edited stem cell therapies for the treatment of diabetes. We believe the combination of ViaCyte’s stem cell capabilities and our gene editing capabilities has the potential to enable a beta-cell replacement product candidate that may deliver durable benefit to patients without requiring concurrent immune suppression.

We have a multi-staged product strategy that leverages our CRISPR/Cas9 platform to advance multiple product candidates incorporating incremental edits designed to increase benefit. Our initial product candidate, VCTX210, is an investigational, allogeneic, gene-edited, immune-evasive, stem cell-derived product candidate for the treatment of type 1 diabetes, or T1D, developed by applying our gene editing technology to ViaCyte’s proprietary stem cell capabilities. VCTX210 has gene edits designed to promote immune evasion and cell fitness. We and ViaCyte are investigating VCTX210 in an ongoing Phase 1 clinical trial that is designed to assess VCTX210’s safety, tolerability, and immune evasion in patients with T1D, and we are in the follow-up stage for this clinical trial. Our next generation product candidate, VCTX211, is an investigational, allogeneic, gene-edited, stem cell-derived product candidate for the treatment of T1D, which incorporates additional gene edits that aim to further enhance cell fitness. In the fourth quarter of 2022, the Clinical Trial Application for VCTX211 was cleared by Health Canada. VCTX211 is being investigated in an ongoing Phase 1/2 clinical trial designed to assess the safety, tolerability and efficacy of VCTX211 in adult patients with T1D.

In Vivo

Our in vivogene editing strategy focuses on gene disruption and whole gene correction – the two technologies required to address the vast majority of the most prevalent severe monogenic diseases as well as many common diseases. We have established a leading platform for in vivogene disruption in the liverediting and are rapidly advancing a broad portfolio of in vivoprograms. Our first in vivo programs target the liver, taking advantage of validated lipid nanoparticle delivery technologies, and aim to treat diseases where we can produce a strong therapeutic effect by safely disrupting a gene with well-understood genetic association. For example, our first two in vivo programs, CTX310 and CTX320, aim to address cardiovascular disease by disrupting the validated targets angiopoietin-like protein 3 and lipoprotein (a), respectively. Phase 1 clinical trials for both CTX310 and CTX320 are ongoing. Building upon CTX310 and CTX320, we have a number of earlier stage investigational in vivo programs leveraging gene disruption in the liver for both rare and common diseases towards clinical trials. Withindiseases. In addition, we have programs focused on gene correction in the liver, we are pursuing diseases that are amenable to a gene disruption strategy and have well-understood genetic linkages, such as cardiovascular disease, or CVD. Our lead investigational in vivoincluding programs CTX310 and CTX320, target angiopoietin-related protein 3 (ANGPTL3) and lipoprotein(a) (Lp(a)), respectively, two validated targets for CVD. We believe this approach of leveraging existing proofs of concept reduces the challenges associated with delivering CRISPR/Cas9-based therapeutics in vivo. Beyond the liver, for delivery to hematopoietic stem cells, the central nervous system, and other extrahepatic tissues,technologies developed by our CRISPR-X research team. Finally, we are pursuing additional delivery technologies, including adeno-associated virus vectors, or AAV, and further advancements to nanoparticle technology.technology and adeno-associated virus, or AAV, vectors, for delivery to tissues beyond the liver, including hematopoietic stem cells.

Type 1 Diabetes

We are advancing a series of programs focused on the development of gene-edited stem cell-derived therapies for the treatment of T1D. We believe our gene editing capabilities have the potential to enable a beta-cell replacement product candidate that may deliver durable benefit to patients without the need for long-term immunosuppression. We have three parallel efforts to achieve this goal. First, our most advanced product candidate, CTX211, is an allogeneic, gene-edited, hypoimmune, stem cell derived product candidate in a device that is implanted into patients and intended to produce insulin in a glucose-dependent manner. This program, formerly known as VCTX211, originated from our collaboration with ViaCyte, Inc., or ViaCyte, a subsidiary of Vertex, and was developed by applying our gene editing technology to ViaCyte’s proprietary stem cell capabilities. CTX211 is being investigated in an ongoing Phase 1/2 clinical trial designed to assess the safety, tolerability and efficacy of CTX211 in adult patients with T1D. Second, we have research efforts focused on a deviceless beta cell replacement approach consisting of unencapsulated beta cells derived from edited stem cells. Third, we have granted a non-exclusive license to certain of our CRISPR/Cas9 intellectual property to Vertex to

18


accelerate Vertex’s development of hypoimmune cell therapies for T1D, for which we received $170 million in upfront and milestone payments in 2023 and remain eligible to receive additional research and development milestones and royalties on future products.

CRISPR-X

While we have made significant progress with our current portfolio of programs, we recognize that we need to continue to innovate to unlock the full potentialpower of CRISPR gene editing and bring the potential ofpotentially transformative therapies to even more patients. In 2022, we launchedWe have a newdedicated early-stage research team known ascalled CRISPR-X that focuses on innovative research to developinnovating next-generation editing modalities. CRISPR-X focuses onis developing technologies to enable whole gene correction and insertion without requiring homology-directed repair or viral delivery of DNA, such as all-RNA gene correction, non-viral delivery of DNA and novel gene insertion techniques.

Partnerships

Given the numerous potential therapeutic applications for CRISPR/Cas9, we have partnered strategically to broaden the indications we can pursue and accelerate development of programs by accessing specific technologies and/or disease-area expertise. We maintain broad partnerships to develop gene editing-based therapeutics in specific disease areas.

Vertex. We established our initial collaboration agreement in 2015 with Vertex, which focused on TDT, SCD, cystic fibrosis and select additional indications. In December 2017, we entered into a joint development and commercialization agreement with Vertex pursuant to which, among other things, we are co-developing and preparing to co-commercialize exa-celco-commercializing CASGEVY for TDT and SCD. In April 2021, we and Vertex amended and restated our existing joint development and commercialization agreement, pursuant to which, among other things, we will continue to develop and prepare to commercialize exa-celCASGEVY for TDT and severe SCD in partnership with Vertex. In addition, weWe also entered into a strategic collaboration and license agreement with Vertex in June 2019 for the development and commercialization of products for the treatment of Duchenne muscular dystrophy, or DMD, and myotonic dystrophy type 1, or DM1 and, in March 2023, we entered into a non-exclusive license agreement with Vertex for Vertex to utilize our gene editing technology in diabetes.

ViaCyte. We entered into a research and collaboration agreement in September 2018 with ViaCyte to pursue the discovery, development and commercialization of gene-edited allogeneic stem cell therapies for the treatment of diabetes, and in July 2021, we entered into a joint development and commercialization agreement with ViaCyte, or the ViaCyte JDCA. In connection with entering into the ViaCyte JDCA, our existing research collaboration agreement with ViaCyte expired in accordance with its terms. Under the ViaCyte JDCA, we and ViaCyte are jointly developing and will commercialize product candidates and shared products for use in the treatment of diabetes type 1, diabetes type 2 and insulin dependent/requiring diabetes, or the ViaCyte Collaboration Field, throughout the world. The ViaCyte JDCA includes, among other things, provisions relating to collaboration and program governance, clinical activities for the product candidates and shared products under the agreement and continuing research by the parties in the ViaCyte

18


Collaboration Field. Unless otherwise mutually agreed, research costs incurred by a party will be solely borne by such party. The program expenses, as originally set forth in the research and collaboration agreement, as applicable, incurred through the date of first commercial sale of a shared product will be allocated 60% to us and 40% to ViaCyte. Following first commercial sale of a shared product, such program expenses will be shared equally between us and ViaCyte. Shared product revenues will be shared equally by us and ViaCyte. In the third quarter of 2022, Vertex announced it had acquired ViaCyte and the rights to the ViaCyte Collaboration Field, and in March 2023, we entered into an amendment to the ViaCyte JDCA pursuant to which, among other things, we adjusted certain rights and obligations of the parties thereunder. In December 2023, ViaCyte elected to opt-out of the collaboration with us for the co-development and co-commercialization of gene-edited stem cell therapies for the treatment of diabetes. Per the opt-out terms, once the opt-out is complete, the on-going collaboration assets will be wholly owned by us, subject to a royalty on future sales owed to ViaCyte. The opt-out became effective in early February 2024. The ViaCyte collaboration assets include CTX211 (formerly VCTX211), an allogeneic, gene-edited, hypoimmune, stem cell derived product candidate in a device that is implanted into patients and intended to produce insulin in a glucose-dependent manner. We are continuing to advance a Phase 1 clinical trial for CTX211 for the treatment of T1D.

Bayer. We entered into an option agreement in the fourth quarter of 2019 with Bayer pursuant to which Bayer has an option to co-develop and co-commercialize two products that we advance for the diagnosis, treatment, or prevention of certain autoimmune disorders, eye disorders, or hemophilia A disorders for a specified period of time, or, under certain circumstances, exclusively license such optioned products.

Other Partnerships. We have entered into a number of additional collaborations and license agreements to support and complement our hematopoietic stem cell, immuno-oncology, regenerative medicine and in vivo programs and platform, including agreements with: Nkarta, Inc. to co-develop and co-commercialize two donor-derived, gene-edited CAR-NK cell product candidates and a product candidate combining NK and T cells; Capsida Biotherapeutics, Inc. to develop in vivo gene editing therapies delivered with engineered AAV vectors for the treatment of amyotrophic lateral sclerosis and Friedreich’s ataxia; Moffitt Cancer Center and Roswell Park Comprehensive Cancer Center to advance autologous CAR T programs against new targets; MaxCyte, Inc. on ex vivo delivery for our hemoglobinopathy and immuno-oncology programs; CureVac AG on optimized mRNA constructs and manufacturing for certain in vivo programs; and KSQ Therapeutics, Inc. on intellectual property for our allogeneic immuno-oncology programs.

Impacts of Coronavirus and Market Conditions on Our Business

We have been actively monitoring the coronavirus pandemic situation and its impact globally. We believe our financial results for the three months ended March 31, 2023 and 2022 were not significantly impacted by the outbreak of the coronavirus. We believe our hybrid and remote working arrangements have had limited impact on our ability to maintain internal operations during such time periods. Further, disruption of global financial markets and a recession or market correction, including as a result of the coronavirus pandemic, the recent failure of certain banks and financial institutions in the United States and globally, the ongoing military conflict between Russia and Ukraine and the related sanctions imposed against Russia, and other global macroeconomic factors such as inflation, could reduce our ability to access capital, which could, in the future, negatively affect our business and the value of our common shares.

Financial Overview

Since our inception in October 2013, we have devoted substantially all of our resources to our research and development efforts, identifying potential product candidates, undertaking drug discovery and preclinical development activities, building and protecting our intellectual property estate, organizing and staffing our company, business planning, raising capital and providing general and administrative support for these operations. To date, we have primarily financed our operations through private placements of our preferred shares, common share issuances, convertible loans and license and collaboration agreements with strategic partners.

19


While we were in a net income position in certain previous years due to certain payments associated with our collaborations with Vertex, we have a history of recurring losses and expect to continue to incur losses for the foreseeable future. Our net losses may fluctuate significantly from quarter to quarter and year to year. We anticipate that our expenses will increase significantly as we continue our current research programs and development activities; seek to identify additional research programs and additional product candidates; conduct initial drug application supporting preclinical studies and initiate clinical trials for our product candidates; initiate preclinical testing and clinical trials for any other product candidates we identify and develop; seek regulatory approval for our product candidates; maintain, defend, protect and expand our intellectual property estate; further develop our gene editing platform; hire additional research, clinical and scientific personnel; incur facilities costs associated with such personnel growth; develop manufacturing infrastructure and conduct related regulatory validation activities; and incur additional costs associated with operating as a public company.

Revenue Recognition

We have not generated any revenue to date from product sales and do not expect to do so in the near future. Revenue recognized for the three months ended March 31, 2024 was not material. Revenue recognized for the three months ended March 31, 2023 was $100.0 million related to our receipt of an upfront payment from Vertex in connection with entering into agreements with Vertex and ViaCyte relating to the research, development, manufacture and commercialization of therapeutic products in the diabetes field. Revenue recognized for the three months ended March 31, 2022 was not material. For additional information about our revenue recognition policy, see Note 2, “Summary of Significant Accounting Policies,” in our Annual Report on Form 10-K for the year ended December 31, 20222023 filed with the SEC on February 21, 2023,2024, as well as Note 6 of the notes to our unaudited condensed consolidated financial statements included in this Quarterly Report on Form 10-Q.

19


Research and Development Expenses

Research and development expenses consist primarily of costs incurred for our research activities, including our product discovery efforts and the development of our product candidates, which include:

employee-related expenses, including salaries, benefits and equity-based compensation expense;
costs of services performed by third parties that conduct research and development and preclinical and clinical activities on our behalf;
costs of purchasing lab supplies and non-capital equipment used in our preclinical activities and in manufacturing preclinical and clinical study materials;
consultant fees;
facility costs, including rent, depreciation and maintenance expenses; and
fees and other payments related to acquiring and maintaining licenses under our third-party licensing agreements.

Our external research and development expenses support our various preclinical and clinical programs, and, as such, we do not break down external research and development expenses further. Our internal research and development expenses consist of payroll and benefits expenses, facilities expense, and other indirect research and development expenses incurred in support of overall research and development activities and, as such, are not allocated to a specific development stage or therapeutic area. Research and development costs are expensed as incurred. Nonrefundable advance payments for research and development goods or services to be received in the future are deferred and capitalized. The capitalized amounts are expensed as the related goods are delivered or the services are performed. At this time, we cannot reasonably estimate or know the nature, timing or estimated costs of the efforts that will be necessary to complete the development of any product candidates we may identify and develop. This is due to the numerous risks and uncertainties associated with developing such product candidates, including the uncertainty of:

successful completion of preclinical studies and IND-enabling studies;
successful enrollment in, and completion of, clinical trials;
receipt of marketing approvals from applicable regulatory authorities;
establishing commercial manufacturing capabilities or making arrangements with third-party manufacturers;
obtaining and maintaining patent and trade secret protection and non-patent exclusivity;
launching commercial sales of the product, if and when approved, whether alone or in collaboration with others;
acceptance of the product, if and when approved, by patients, the medical community and third-party payors;
effectively competing with other therapies and treatment options;
a continued acceptable safety profile following approval;
enforcing and defending intellectual property and proprietary rights and claims; and
achieving desirable medicinal properties for the intended indications.

20


A change in the outcome of any of these variables with respect to the development of any product candidates or the subsequent commercialization of any product candidates we may successfully develop could significantly change the costs, timing and viability associated with the development of that product candidate.

Research and development activities are central to our business model. We expect to continue to incur research and development costs consistent with research and development at companies of our size and stage of development, which may increase in the foreseeable future as our current development programs progress, new programs are added and we continue to prepare regulatory filings. These increases will likely include the costs related to the implementation and expansion of clinical trial sites and related patient enrollment, monitoring, program management and manufacturing expenses for current and future clinical trials.

General and Administrative Expenses

General and administrative expenses consist primarily of employee related expenses, including salaries, benefits and equity-based compensation, for personnel in executive, finance, accounting, business development, human resources and other general and administrative functions. Other significant costs include facility costs not otherwise included in research and development expenses, legal fees relating to patent and corporate matters and fees for accounting and consulting services.

We expect to continue to incur general and administrative expenses consistent with general and administrative functions at research and development at companies of our size and stage of development, which may increase in the future to support continued research and development activities, and potential commercialization of our product candidates. In addition, we anticipate ongoing expenses related to the reimbursements of third-party patent related expenses in connection with certain of our in-licensed intellectual property.

20


Collaboration Expense, Net

Collaboration expense, net, consists of operating expense related to the exa-celCASGEVY program under our collaboration with Vertex. Under the A&R Vertex JDCA, we have an option to defer our portion of specified costs on the exa-celCASGEVY program in excess of $110.3 million for the years ended December 31, 2022, 2023 and 2024. Vertex mayAny deferred amounts are only recover any such deferred amountspayable to Vertex as an offset against future profitability of the exa-celCASGEVY program determined on an annual basis in accordance withand the A&R Vertex JDCA. Any such deferred amounts payable are capped at a specified maximum amount per year.

Other Income (Expense), Net

Other income (expense), net consists primarily of interest income earned on investments.

Results of Operations

Comparison of three months ended March 31, 20232024 and 20222023 (in thousands):

 

Three Months Ended March 31,

 

 

Period to Period

 

 

Three Months Ended March 31,

 

 

Period to Period

 

 

2023

 

 

2022

 

 

Change

 

 

2024

 

 

2023

 

 

Change

 

Revenue:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Collaboration revenue

 

$

100,000

 

 

$

178

 

 

$

99,822

 

 

$

 

 

$

100,000

 

 

$

(100,000

)

Grant revenue

 

 

 

 

 

762

 

 

 

(762

)

 

 

504

 

 

 

 

 

 

504

 

Total revenue

 

 

100,000

 

 

 

940

 

 

 

99,060

 

 

 

504

 

 

 

100,000

 

 

 

(99,496

)

Operating expenses:

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Research and development

 

 

99,935

 

 

 

118,245

 

 

 

(18,310

)

 

 

76,172

 

 

 

99,935

 

 

 

(23,763

)

General and administrative

 

 

22,360

 

 

 

28,021

 

 

 

(5,661

)

 

 

17,953

 

 

 

22,360

 

 

 

(4,407

)

Collaboration expense, net

 

 

42,192

 

 

 

30,646

 

 

 

11,546

 

 

 

46,966

 

 

 

42,192

 

 

 

4,774

 

Total operating expenses

 

 

164,487

 

 

 

176,912

 

 

 

(12,425

)

 

 

141,091

 

 

 

164,487

 

 

 

(23,396

)

Loss from operations

 

 

(64,487

)

 

 

(175,972

)

 

 

111,485

 

 

 

(140,587

)

 

 

(64,487

)

 

 

(76,100

)

Other income, net

 

 

12,742

 

 

 

363

 

 

 

12,379

 

 

 

24,720

 

 

 

12,742

 

 

 

11,978

 

Loss before income taxes

 

 

(51,745

)

 

 

(175,609

)

 

 

123,864

 

 

 

(115,867

)

 

 

(51,745

)

 

 

(64,122

)

Benefit for income taxes

 

 

(1,320

)

 

 

(3,608

)

 

 

2,288

 

Provision (benefit) for income taxes

 

 

(724

)

 

 

(1,320

)

 

 

596

 

Net loss

 

$

(53,065

)

 

$

(179,217

)

 

$

126,152

 

 

$

(116,591

)

 

$

(53,065

)

 

$

(63,526

)

Collaboration Revenue

There was no collaboration revenue for the three months ended March 31, 2024. Collaboration revenue for the three months ended March 31, 2023 was $100.0 million due to an upfront payment from Vertex. Revenue for the three months ended March 31, 2022 was not material. Please refer to Note 6 of the notes to our unaudited condensed consolidated financial statements included in this Quarterly Report on Form 10-Q for further information.

21


Research and Development Expenses

Research and development expenses were $76.2 million for the three months ended March 31, 2024, compared to $99.9 million for the three months ended March 31, 2023, compared to $118.2 million for the three months ended March 31, 2022.2023. The following table summarizes our research and development expenses for the three months ended March 31, 20232024 and 2022,2023, together with the changes in those items in dollars (in thousands):

 

Three Months Ended March 31,

 

 

Period to Period

 

 

Three Months Ended March 31,

 

 

Period to Period

 

 

2023

 

 

2022

 

 

Change

 

 

2024

 

 

2023

 

 

Change

 

External research and development expenses

 

$

34,493

 

 

$

45,476

 

 

$

(10,983

)

 

$

18,669

 

 

$

34,493

 

 

$

(15,824

)

Employee related expenses

 

 

22,558

 

 

 

21,191

 

 

 

1,367

 

 

 

20,991

 

 

 

22,558

 

 

 

(1,567

)

Facility expenses

 

 

28,400

 

 

 

30,780

 

 

 

(2,380

)

 

 

22,769

 

 

 

28,400

 

 

 

(5,631

)

Stock-based compensation expenses

 

 

11,676

 

 

 

14,589

 

 

 

(2,913

)

 

 

11,182

 

 

 

11,676

 

 

 

(494

)

Other expenses

 

 

693

 

 

 

539

 

 

 

154

 

 

 

369

 

 

 

693

 

 

 

(324

)

Sublicense and license fees

 

 

2,115

 

 

 

5,670

 

 

 

(3,555

)

 

 

2,192

 

 

 

2,115

 

 

 

77

 

Total research and development expenses

 

$

99,935

 

 

$

118,245

 

 

$

(18,310

)

 

$

76,172

 

 

$

99,935

 

 

$

(23,763

)

The decrease of approximately $18.3$23.8 million was primarily attributable to the following:

$11.015.8 million of decreased external research and development costs, primarily associated with a decrease in variable external research and manufacturing costs; and
$3.65.6 million of decreased sublicense and license fees; andfacility-related expenses, primarily driven by lower lab consumable costs.

21General and Administrative Expenses


General and administrative expenses were $18.0 million for the three months ended March 31, 2024, compared to general and administrative expenses of $22.4 million for the three months ended March 31, 2023. The decrease in general and administrative expenses of approximately $4.4 million was primarily attributable to the following:

$2.9 2.4 million of decreased employee related and stock-based compensation expensesexpense primarily due to an overall decrease in the fair value of equity awards granted in 20222023 and into 20232024; and subsequent reduction in stock-based compensation expense incurred.

General and Administrative Expenses

General and administrative expenses were $22.4

$1.8 million for the three months ended March 31, 2023, compared to general and administrative expenses of $28.0 million for the three months ended March 31, 2022. The decrease in general and administrative expenses of $5.6 million was primarily attributable to decreased stock-based compensation expense and decreased intellectual propertyexternal professional costs.

Collaboration Expense, Net

Collaboration expense, net, was $47.0 million for the three months ended March 31, 2024, compared to $42.2 million for the three months ended March 31, 2023, compared to $30.6 million for the three months ended March 31, 2022.2023. The increase of approximately $11.5$4.8 million in collaboration expense, net, was primarily attributable to increasedcommercial and manufacturing and other pre-commercial costs with regards to the exa-cel program.costs.

Other Income, Net

Other income was $12.7$24.7 million for the three months ended March 31, 2023,2024, compared to $0.4$12.7 million of income for the three months ended March 31, 2022.2023. The increase of approximately $12.0 million was primarily due to interest income earned on cash, cash equivalents and marketable securities for the three months ended March 31, 2023.2024.

Liquidity and Capital Resources

We have predominantly incurred losses and cumulative negative cash flows from operations since our inception. As of March 31, 2023,2024, we had $1,889.5$2,108.1 million in cash, cash equivalents and marketable securities, of which approximately $2.5$1.8 million was held outside of the United States, and an accumulated deficit of $899.2$1,116.3 million. We anticipate that we will continue to incur losses for at least the next several years. We expect to continue to incur research and development costs and general and administrative expenses consistent with costs associated with research and development at companies of our size and stage of development, and, as a result, we will need additional capital to fund our operations, which we may raise through public or private equity or debt financings, strategic collaborations, or other sources.

In August 2019, we entered into the August 2019 Sales Agreement with Jefferies and filed our current prospectus supplement for $419.8 million in July 2021. As of March 31, 2023,2024, we have issued and sold an aggregate of 1.11.5 million common shares under the current prospectus supplement at an average price of $168.79$139.28 per share for aggregate proceeds of $178.8$212.4 million, which were net of equity issuance costs of $2.4$2.9 million.

22


In February 2024, the Company entered into an investment agreement for the sale of approximately $280.0 million of its common shares to a group of institutional investors in a registered direct offering, at a price per share of $71.50. The Company received net proceeds of $279.0 million, excluding stamp taxes due of $2.8 million.

Funding Requirements

Our primary uses of capital are, and we expect will continue to be, research and development activities, manufacturing activities, compensation and related expenses, laboratory and related supplies, legal and other regulatory expenses, patent prosecution, filing, defense and intellectual property maintenance costs, and general overhead costs, including costs associated with operating as a public company. We expect to continue to incur operating expenses consistent with costs associated with research and development at companies of our size and stage of development, which may increase in the future to support continued research and development activities and potential commercialization of our product candidates.

Because most of our programs are still in early stages of development and the outcome of these efforts is uncertain, we cannot estimate the actual amounts necessary to successfully complete the development, manufacture and commercialization of any current or future product candidates, if approved, or whether, or when, we may achieve profitability. Until such time as we can generate substantial product revenues, if ever, we expect to finance our cash needs through a combination of equity financings, debt financings and payments received in connection with our collaboration agreements. We intend to consider opportunities to raise additional funds through the sale of equity or debt securities when market conditions are favorable to us to do so. However, the trading prices for our common shares and other biopharmaceutical companies have been highly volatile. As a result, we may face difficulties raising capital through sales of our common shares or such sales may be on unfavorable terms. In addition, a recession, depression or other sustained adverse market event, including resulting from the continued spread of the coronavirus or the recent failure of certain banks and financial institutions in the United States and globally, could materially and adversely affect our business and the value of our common shares. To the extent that we raise additional capital through the future sale of equity or debt securities, the ownership interests of our shareholders will be diluted, and the terms of these securities may include liquidation or other preferences that adversely affect the rights of our existing shareholders. If we raise additional funds through collaboration arrangements in the future, we may have to relinquish valuable rights to our technologies, future revenue streams or product candidates or grant licenses on terms that may not be favorable to us. If we are unable to raise additional funds through equity or debt financings when needed, we may be

22


required to delay, limit, reduce or terminate our product development or future commercialization efforts or grant rights to develop and market product candidates that we would otherwise prefer to develop and market ourselves.

Outlook

Based on our research and development plans and our timing expectations related to the progress of our programs, we expect our existing cash will enable us to fund our operating expenses and capital expenditures for at least the next 24 months without giving effect to any additional proceeds we may receive under our collaboration with Vertex and any other capital raising transactions we may complete. We have based this estimate on assumptions that may prove to be wrong, and we could use our capital resources sooner than we expect. Given our need for additional financing to support the long-term clinical development of our programs, we intend to consider additional financing opportunities when market terms are favorable to us.

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

23


Cash Flows

The following table provides information regarding our cash flows for each of the periods below (in thousands):

 

 

Three Months Ended March 31,

 

 

Period to Period

 

 

 

2023

 

 

2022

 

 

Change

 

Net cash provided by (used in) operating activities

 

$

8,799

 

 

$

(135,239

)

 

$

144,038

 

Net cash provided by (used in) investing activities

 

 

117,829

 

 

 

(114,515

)

 

 

232,344

 

Net cash provided by financing activities

 

 

5,404

 

 

 

10,649

 

 

 

(5,245

)

Effect of exchange rate changes on cash

 

 

32

 

 

 

(27

)

 

 

59

 

Net increase (decrease) in cash

 

$

132,064

 

 

$

(239,132

)

 

$

371,196

 

 

 

Three Months Ended March 31,

 

 

Period to Period

 

 

 

2024

 

 

2023

 

 

Change

 

Net cash provided by operating activities

 

$

109,752

 

 

$

8,799

 

 

$

100,953

 

Net cash (used in) provided by investing activities

 

 

(97,800

)

 

 

117,829

 

 

 

(215,629

)

Net cash provided by financing activities

 

 

305,934

 

 

 

5,404

 

 

 

300,530

 

Effect of exchange rate changes on cash

 

 

(11

)

 

 

32

 

 

 

(43

)

Net increase in cash

 

$

317,875

 

 

$

132,064

 

 

$

185,811

 

Operating Activities

Net cash provided by operating activities was $109.8 million for the three months ended March 31, 2024, compared to cash provided by in operating activities of $8.8 million for the three months ended March 31, 2023, compared to cash used in operating activities of $135.2 million for the three months ended March 31, 2022.2023. The increase from a cash used in operating activities position to anet cash provided by operating activities position of $144.0approximately $101.0 million was primarily driven by a reductionan increase in net changes of operating assets and liabilities by $173.6 million, while non-cash expense decreased by $9.1 million. Additionally, the increase was offset by an increase in our net loss position of $126.2$63.5 million, from a net loss of $179.2 million for the three months ended March 31, 2022 to net loss of $53.1 million for the three months ended March 31, 2023 to a net loss of $116.6 million for the three months ended March 31, 2024 driven by revenue recognized in the connection with an upfront payment from Vertex in the first quarter of 2023. Additionally, non-cash expense decreased by $12.9 million, primarily related to a decrease2023 that did not recur in stock-based compensation expense and amortizationthe first quarter of premiums and discounts on marketable securities, while net changes of operating assets and liabilities increased $30.8 million.2024.

Investing Activities

Net cash provided byused in investing activities for the three months ended March 31, 20232024 was $117.8$97.8 million, compared to net cash used inprovided by investing activities of $114.5$117.8 million for the three months ended March 31, 2022.2023. The change from a net cash used in investing activities to a net cash provided by investing activities consistedwas primarily of andriven by a net increase in purchases of our marketable securities, maturities, in addition tooffset by a reduction of purchases of marketable securities and property and equipment.equipment purchases.

Financing Activities

Net cash provided by financing activities for the three months ended March 31, 20232024 was $5.4$305.9 million, compared with $10.6$5.4 million for the three months ended March 31, 2022.2023. Net cash provided by financing activities for the three months ended March 31, 2024 and 2023 and 2022consisted of proceeds from the sale of approximately $280.0 million of its common shares to a group of institutional investors in a registered direct offering, at a price per share of $71.50. The Company received net proceeds of $279.0 million, excluding stamp taxes due of $2.8 million. Additionally, net cash provided by financing activities consisted of option exercise proceeds, net of issuance costs.

23


Critical Accounting Policies and Significant Judgments and Estimates

This discussion and analysis of our financial condition and results of operations is based on our financial statements, which we have prepared in accordance with U.S. GAAP. We believe that several accounting policies are important to understanding our historical and future performance. We refer to these policies as critical because these specific areas generally require us to make judgments and estimates about matters that are uncertain at the time we make the estimate, and different estimates—which also would have been reasonable—could have been used. On an ongoing basis, we evaluate our estimates and judgments, including those described in greater detail below. We base our estimates on historical experience and other market-specific or other relevant assumptions that we believe to be reasonable under the circumstances, the results of which form the basis for making judgments about the carrying value of assets and liabilities that are not readily apparent from other sources. Actual results may differ from these estimates under different assumptions or conditions.

We believe that our most critical accounting policies are those relating to revenue recognition and equity-based compensation, and there have been no changes to our accounting policies discussed in our Annual Report on Form 10-K for the year ended December 31, 20222023 filed with the SEC on February 21, 2023.2024.

Recent Accounting Pronouncements

Refer to Note 1 of the notes to our unaudited condensed consolidated financial statements included in this Quarterly Report on Form 10-Q for a discussion of recent accounting pronouncements.

24


Item 3. Qualitative and Quantitative Disclosures about Market Risk

Interest Rate Sensitivity

We are exposed to market risk related to changes in interest rates. As of March 31, 2023,2024, we had cash, cash equivalents and marketable securities of $1,889.5$2,108.1 million, primarily invested in U.S. treasury securities and government agency securities, corporate bonds, commercial paper and money market accounts invested in U.S. government agency securities. Due to the conservative nature of these instruments, we do not believe that we have a material exposure to interest rate risk. If interest rates were to increase or decrease by 1%, the fair value of our investment portfolio would increase or decrease by an immaterial amount.

Foreign Currency Exchange Rate Risk

As a result of our foreign operations, we face exposure to movements in foreign currency exchange rates, primarily the Swiss Franc and British Pound, against the U.S. dollar. The current exposures arise primarily from cash, accounts payable and intercompany receivables and payables. Changes in foreign exchange rates affect our consolidated statement of operations and distort comparisons between periods. To date, foreign currency transaction gains and losses have not been material to our financial statements, and we have not engaged in any foreign currency hedging transactions.

Inflation

Inflation generally affects us by increasing our cost of labor, clinical trial and manufacturing costs. We do not believe that inflation had a material effect on our business, financial condition or results of operations during the three months ended March 31, 20232024 and 2022.2023.

24


Item 4. Controls and Procedures.

Management’s Evaluation of our Disclosure Controls and Procedures

We maintain disclosure controls and procedures that are designed to ensure that information required to be disclosed in the reports that we file or submit under the Securities and Exchange Act of 1934 is (1) recorded, processed, summarized, and reported within the time periods specified in the SEC’s rules and forms and (2) accumulated and communicated to our management, including our principal executive officer and principal financial officer, to allow timely decisions regarding required disclosure.

As of March 31, 2023,2024, our management, with the participation of our principal executive officer and principal financial officer, evaluated the effectiveness of our disclosure controls and procedures (as defined in Rules 13a-15(e) and 15d-15(e) under the Securities and Exchange Act of 1934). Our 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. Our principal executive officer and principal financial officer have concluded based upon the evaluation described above that, as of March 31, 2023,2024, our disclosure controls and procedures were effective at the reasonable assurance level.

Changes in Internal Control over Financial Reporting

During the quarter ended March 31, 2023,2024, there were no changes in our internal control over financial reporting, as such term is defined in Rules 13a-15(f) and 15(d)-15(f) promulgated under the Securities Exchange Act of 1934, that have materially affected, or are reasonably likely to materially affect, our internal control over financial reporting.

25


 

PART II—OTHER INFORMATION

In the ordinary course of business, we are from time to time involved in lawsuits, investigations, proceedings and threats of litigation related to, among other things, our intellectual property estate (including certain in-licensed intellectual property), commercial arrangements and other matters. Such proceedings may include quasi-litigation, inter partes administrative proceedings in the U.S. Patent and Trademark Office and the European Patent Office involving our intellectual property estate including certain in-licensed intellectual property. There are currently no claims or actions pending against us that, in the opinion of our management, are likely to have a material adverse effect on our business.

There have been no material developments with respect to the legal proceedings previously disclosed in “Item 3. Legal Proceedings” of our Annual Report on Form 10-K for the fiscal year ended December 31, 20222023 filed with the SEC on February 21, 2023.2024.

Item 1A. Risk Factors.

We are updating and supplementing our risk factors previously disclosed in Item 1A of our Annual Report on Form 10-K for the year ended December 31, 2022 filed with the SEC on February 21, 2023 to add the following new risk factor:

Conditions in the banking system and financial markets, including the failure of banks and financial institutions, could have an adverse effect on our operations and financial results.

Actual events involving limited liquidity, defaults, non-performance or other adverse developments that affect financial institutions, transactional counterparties or other companies in the financial services industry or the financial services industry generally, or concerns or rumors about any events of these kinds or other similar risks, have in the past and may in the future lead to market-wide liquidity problems. For example, on March 10 and March 12, 2023, the Federal Deposit Insurance Corporation took control and was appointed receiver of Silicon Valley Bank, or SVB, and Signature Bank and Silvergate Capital Corp., or Silvergate Capital, respectively, after each bank was unable to continue their operations. Since then, additional financial institutions have experienced similar failures and have been placed into receivership. It is possible that other banks will face similar difficulty in the future. These events exposed vulnerabilities in the banking sector, including legal uncertainties, significant volatility and contagion risk, and caused market prices of regional bank stocks to plummet.

As of the date of this Quarterly Report on Form 10-Q, our exposure to SVB, Signature Bank, Silvergate Capital, and other banks in financial difficulty is immaterial; however, we are unable to predict the extent or nature of the impacts of these evolving circumstances at this time. If, for example, other banks and financial institutions enter receivership or become insolvent in the future in response to financial conditions affecting the banking system and financial markets, our ability to access our existing cash, cash equivalents and investments may be threatened. While it is not possible at this time to predict the extent of the impact that high market volatility and instability of the banking sector could have on economic activity and our business in particular, the failure of other banks and financial institutions and the measures taken by governments, businesses and other organizations in response to these events could adversely impact our business, financial condition and results of operations.

There have been no other material changes to the risk factors previously disclosed in Part I, Item 1A (Risk Factors) of our Annual Report on Form 10-K for the year ended December 31, 2022. Please refer to the complete Part I, Item 1A of our Annual Report for additional risks and uncertainties we are facing that may have a material adverse effect on our business prospects, financial condition and results of operations. In addition to the risks described in our Annual Report on Form 10-K, you should carefully consider the other information set forth in this Form 10-Q and the information in our other filings with the SEC, as they could materially affect our business, financial condition or future results of operations. There have been no material changes to the risk factors previously disclosed in Part I, Item 1A (Risk Factors) of our Annual Report on Form 10-K.

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

None.

Item 3. Defaults Upon Senior Securities.

Not applicable.

Item 4. Mine Safety Disclosures.

Not applicable.

26


Item 5. Other Information.

None.From time to time, our officers (as defined in Rule 16a–1(f)) and directors may enter into Rule 10b5-1 or non-Rule 10b5-1 trading arrangements (as each such term is defined in Item 408 of Regulation S-K). During the three months ended March 31, 2024, our officers and directors took the following actions with respect to 10b5-1 trading arrangements:

On March 4, 2024, Jim Kasinger, our General Counsel and Secretary, adopted a Rule 10b5-1 trading arrangement with respect to the sale of up to an aggregate of 93,632 common shares of the Company pursuant to the terms of such trading plan. Mr. Kasinger's Rule 10b5-1 trading arrangement is active through June 30, 2025.

26


Item 6. Exhibits

The exhibits filed as part of this Quarterly Report on Form 10-Q are set forth in the Exhibit Index below.

 

Exhibit

Number

 

Description of Document

10.1

Employment Agreement, dated March 14, 2023, by and between CRISPR Therapeutics, Inc., and Raju Prasad, Ph.D. (incorporated herein by reference to Exhibit 10.1 to the Company's Current Report on Form 8-K filed on March 14, 2023).

10.2†^

Non-Exclusive License Agreement, dated March 23, 2023, by and between Vertex Pharmaceuticals Incorporated and CRISPR Therapeutics AG (incorporated herein by reference to Exhibit 10.1 to the Company’s Current Report on Form 8-K filed on March 27, 2023).

 

 

 

31.1*

 

Certification of Chief Executive Officer Pursuant to Section 302 of the Sarbanes-Oxley Act of 2002

 

 

 

31.2*

 

Certification of Chief Financial Officer Pursuant to Section 302 of the Sarbanes-Oxley Act of 2002

 

 

 

32.1*+

 

Certification 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 instance document 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 (formatted as inline XBRL with applicable taxonomy extension information contained in Exhibits 101)

 

* Filed herewith.

† Certain portions of this exhibit have been omitted because they are not material and the registrant customarily and actually treats that information as private or confidential.

^ Certain exhibits and schedules to these agreements have been omitted pursuant to Item 601 of Regulation S-K. The registrant will furnish copies of any of the exhibits and schedules to the Securities and Exchange Commission upon request.

+ The certification attached as Exhibit 32.1 that accompany this Quarterly Report on Form 10-Q are not deemed filed with the Securities and Exchange Commission and are not to be incorporated by reference into any filing of CRISPR Therapeutics AG under the Securities Act of 1933, as amended, or the Securities Exchange Act of 1934, as amended, whether made before or after the date of this Form 10-Q, irrespective of any general incorporation language contained in such filing.

27


 

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.

 

CRISPR Therapeutics AG

 

 

 

Dated: May 8, 20232024

By:

/s/ Samarth Kulkarni

 

Samarth Kulkarni

 

Chief Executive Officer

 

(Principal Executive Officer)

 

 

 

Dated: May 8, 20232024

By:

/s/ Raju Prasad

 

Raju Prasad

 

Chief Financial Officer

 

(Principal Financial Officer)

 

28