UNITED STATES
SECURITIES AND EXCHANGE COMMISSION
Washington, DC 20549

FORM 10-Q

(Mark One)
xQUARTERLY REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934
For the quarterly period ended June 30, 20222023
oTRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934
For the transition period from          to          .
Commission file number 001-38042

ARROWHEAD PHARMACEUTICALS, INC.
(Exact name of registrant as specified in its charter)

Delaware46-0408024
(State or other jurisdiction of incorporation or organization)(I.R.S. Employer Identification No.)
177 E. Colorado Blvd, Suite 700
Pasadena, California 91105
(626) 304-3400
(Address and telephone number of principal executive offices)
Former name, former address, and former fiscal year, if changed since last report: N/A

Securities registered pursuant to Section 12(b) of the Exchange Act:
Title of each classTrading Symbol(s)Name of each exchange on which registered
Common Stock, par value $0.001 per shareARWRThe Nasdaq Global Select Market
Indicate by check mark whether the registrant (1) has filed all reports required to be filed by Section 13 or 15(d) of the Securities Exchange Act of 1934 during the preceding 12 months (or for such shorter period that the registrant was required to file such reports), and (2) has been subject to such filing requirements for the past 90 days. Yes x No o
Indicate by check mark whether the registrant has submitted electronically every Interactive Data File required to be submitted pursuant to Rule 405 of Regulation S-T (§ 232.405 of this chapter) during the preceding 12 months (or for such shorter period that the registrant was required to submit such files). Yes x No o
Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, a smaller reporting company, or an emerging growth company. See the definitions of “large accelerated filer,” “accelerated filer,” “smaller reporting company” and “emerging growth company” in Rule 12b-2 of the Exchange Act.
Large Accelerated FilerxAccelerated Filero
Non-Accelerated FileroSmaller Reporting Companyo
Emerging Growth Companyo
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. o
Indicate by check mark whether the registrant is a shell company (as defined in Rule 12b-2 of the Exchange Act). Yes o No x
The number of shares of the registrant’s common stock outstanding as of July 28, 202231, 2023 was 105,848,963.107,192,901.



Page(s)



PART I. FINANCIAL INFORMATION
ITEM 1.    FINANCIAL STATEMENTS
Arrowhead Pharmaceuticals, Inc.
Consolidated Balance Sheets
(In thousands, except per share amounts)
June 30, 2023September 30, 2022
(unaudited) June 30, 2022September 30, 2021(unaudited)
ASSETSASSETSASSETS
CURRENT ASSETS
Cash and cash equivalents$139,439 $184,434 
Current assets:Current assets:
Cash, cash equivalents and restricted cashCash, cash equivalents and restricted cash$105,334 $108,005 
Accounts receivableAccounts receivable239 10,255 Accounts receivable1,247 1,410 
Short term investmentsShort term investments346,369 268,391 
Prepaid expensesPrepaid expenses6,761 4,362 Prepaid expenses10,053 7,289 
Other current assetsOther current assets8,892 2,191 Other current assets7,162 20,204 
Marketable securities— 126,728 
Short term investments277,057 56,627 
TOTAL CURRENT ASSETS432,388 384,597 
Total current assetsTotal current assets470,165 405,299 
Property and equipment, netProperty and equipment, net71,904 48,675 Property and equipment, net231,369 110,297 
Intangible assets, netIntangible assets, net12,387 13,663 Intangible assets, net10,687 11,962 
Long term investments165,920 245,595 
Long-term investmentsLong-term investments42,758 105,872 
Right-of-use assetsRight-of-use assets68,908 17,346 Right-of-use assets40,667 58,291 
Other assetsOther assets275 272 Other assets210 218 
TOTAL ASSETS$751,782 $710,148 
Total AssetsTotal Assets$795,856 $691,939 
LIABILITIES, NONCONTROLLING INTEREST AND STOCKHOLDERS’ EQUITYLIABILITIES, NONCONTROLLING INTEREST AND STOCKHOLDERS’ EQUITYLIABILITIES, NONCONTROLLING INTEREST AND STOCKHOLDERS’ EQUITY
CURRENT LIABILITIES
Current liabilities:Current liabilities:
Accounts payableAccounts payable$5,894 $9,457 Accounts payable$7,874 $2,868 
Accrued expensesAccrued expenses32,499 14,001 Accrued expenses38,191 46,856 
Accrued payroll and benefitsAccrued payroll and benefits2,648 9,773 Accrued payroll and benefits4,434 12,251 
Lease liabilitiesLease liabilities2,883 2,250 Lease liabilities2,823 2,776 
Deferred revenueDeferred revenue84,288 111,055 Deferred revenue16,905 74,099 
TOTAL CURRENT LIABILITIES128,212 146,536 
LONG-TERM LIABILITIES
Total current liabilitiesTotal current liabilities70,227 138,850 
Long-term liabilities:Long-term liabilities:
Lease liabilities, net of current portionLease liabilities, net of current portion78,231 23,295 Lease liabilities, net of current portion79,911 78,800 
Deferred revenue, net of current portionDeferred revenue, net of current portion71,162 131,495 Deferred revenue, net of current portion— 55,950 
TOTAL LONG-TERM LIABILITIES149,393 154,790 
Liability related to the sale of future royaltiesLiability related to the sale of future royalties263,064 — 
Other liabilitiesOther liabilities669 — 
Total long-term liabilitiesTotal long-term liabilities343,644 134,750 
Commitments and contingencies (Note 7)Commitments and contingencies (Note 7)00Commitments and contingencies (Note 7)
STOCKHOLDERS’ EQUITY
Arrowhead Pharmaceuticals, Inc. stockholders’ equity:
Common stock, $0.001 par value; 145,000 shares authorized; 105,795 and 104,327 shares issued and outstanding as of June 30, 2022 and September 30, 2021, respectively198 197 
Noncontrolling interest and stockholders’ equity:Noncontrolling interest and stockholders’ equity:
Common stock, $0.001 par value:
Authorized 290,000 and 145,000 shares; issued and outstanding 107,102 and 105,960 shares
Common stock, $0.001 par value:
Authorized 290,000 and 145,000 shares; issued and outstanding 107,102 and 105,960 shares
199 198 
Additional paid-in capitalAdditional paid-in capital1,189,113 1,053,386 Additional paid-in capital1,281,393 1,219,213 
Accumulated other comprehensive lossAccumulated other comprehensive loss(140)(69)Accumulated other comprehensive loss(411)(136)
Accumulated deficitAccumulated deficit(735,244)(644,692)Accumulated deficit(916,351)(820,755)
TOTAL ARROWHEAD PHARMACEUTICALS INC. STOCKHOLDERS' EQUITY453,927 408,822 
Noncontrolling Interest20,250 — 
TOTAL STOCKHOLDERS’ EQUITY474,177 408,822 
TOTAL LIABILITIES, NONCONTROLLING INTEREST AND STOCKHOLDERS’ EQUITY$751,782 $710,148 
Total Arrowhead Pharmaceuticals, Inc. stockholders’ equityTotal Arrowhead Pharmaceuticals, Inc. stockholders’ equity364,830 398,520 
Noncontrolling interestNoncontrolling interest17,155 19,819 
Total noncontrolling interest and stockholders’ equityTotal noncontrolling interest and stockholders’ equity381,985 418,339 
Total Liabilities, Noncontrolling Interest and Stockholders’ EquityTotal Liabilities, Noncontrolling Interest and Stockholders’ Equity$795,856 $691,939 
The accompanying notes are an integral part of these unaudited consolidated financial statements.
1


Arrowhead Pharmaceuticals, Inc.
Consolidated Statements of Operations and Comprehensive Income (Loss)
(unaudited)Loss
(In thousands, except per share amounts)
Three Months Ended June 30,Nine Months Ended June 30,
2022202120222021
REVENUE$32,412 $45,891 $211,656 $100,004 
OPERATING EXPENSES
Research and development72,180 59,325 213,930 140,576 
General and administrative expenses33,141 18,434 92,403 43,581 
TOTAL OPERATING EXPENSES105,321 77,759 306,333 184,157 
OPERATING INCOME (LOSS)(72,909)(31,868)(94,677)(84,153)
OTHER INCOME
Interest income, net1,240 1,280 3,450 4,972 
Other income (expense)(377)664 675 1,707 
TOTAL OTHER INCOME863 1,944 4,125 6,679 
INCOME (LOSS) BEFORE INCOME TAXES(72,046)(29,924)(90,552)(77,474)
Provision for income taxes— — — — 
NET INCOME (LOSS)(72,046)(29,924)(90,552)(77,474)
NET INCOME (LOSS) PER SHARE - BASIC$(0.68)$(0.29)$(0.86)$(0.75)
NET INCOME (LOSS) PER SHARE - DILUTED$(0.68)$(0.29)$(0.86)$(0.75)
Weighted average shares outstanding - basic105,753 104,099 105,273 103,569 
Weighted average shares outstanding - diluted105,753 104,099 105,273 103,569 
OTHER COMPREHENSIVE INCOME (LOSS), NET OF TAX:
Foreign currency translation adjustments(33)(40)(71)44 
COMPREHENSIVE INCOME (LOSS)$(72,079)$(29,964)$(90,623)$(77,430)
(unaudited)
Three Months Ended June 30,Nine Months Ended June 30,
2023202220232022
Revenue$15,825 $32,412 $224,638 $211,656 
Operating expenses:
Research and development94,757 72,180 253,333 213,930 
General and administrative23,771 33,141 67,977 92,403 
Total operating expenses118,528 105,321 321,310 306,333 
Operating loss(102,703)(72,909)(96,672)(94,677)
Other income (expense):
Interest income4,172 1,240 11,414 3,450 
Interest expense(5,158)— (13,064)— 
Other, net306 (377)821 675 
Total other (loss) income(680)863 (829)4,125 
Loss before income tax expense and noncontrolling interest(103,383)(72,046)(97,501)(90,552)
Income tax expense742 — 759 — 
Net loss including noncontrolling interest(104,125)(72,046)(98,260)(90,552)
Net loss attributable to noncontrolling interest, net of tax(1,179)— (2,664)— 
Net loss attributable to Arrowhead Pharmaceuticals, Inc.$(102,946)$(72,046)$(95,596)$(90,552)
Net loss per share attributable to Arrowhead Pharmaceuticals, Inc.:
Basic$(0.96)$(0.68)$(0.90)$(0.86)
Diluted$(0.96)$(0.68)$(0.90)$(0.86)
Weighted-average shares used in calculating
Basic107,004 105,753 106,597 105,273 
Diluted107,004 105,753 106,597 105,273 
Other comprehensive loss, net of tax:
Foreign currency translation adjustments(79)(33)(275)(71)
Comprehensive loss$(104,204)$(72,079)$(98,535)$(90,623)
The accompanying notes are an integral part of these unaudited consolidated financial statements.
2


Arrowhead Pharmaceuticals, Inc.
Consolidated Statements of Stockholders’ Equity and Noncontrolling Interest
(in thousands)
(unaudited)
(In thousands, except per share amounts)
Common
Stock
Amount ($)
Additional
Paid-In
Capital
Accumulated
Other
Comprehensive
Loss
Accumulated
Deficit
Non-
controlling Interest
Totals
Balance at September 30, 2022105,960 $198 $1,219,213 $(136)$(820,755)$19,819 $418,339 
Stock-based compensation— — 19,390 — — — 19,390 
Exercise of stock options82 — 576 — — — 576 
Common stock - restricted stock units vesting98 (1)— — —  
Foreign currency translation adjustments— — — (122)— — (122)
Interest in joint venture— — — — — (486)(486)
Net loss for the three months ended December 31, 2022— — — — (41,325)— (41,325)
Balance at December 31, 2022106,140 $199 $1,239,178 $(258)$(862,080)$19,333 $396,372 
Stock-based compensation— — 20,612 — — — 20,612 
Exercise of stock options64 — 520 — — — 520 
Common stock - restricted stock units vesting665 — — — — —  
Foreign currency translation adjustments— — — (74)— — (74)
Interest in joint venture— — — — — (999)(999)
Net income for the three months ended March 31, 2023— — — — 48,675 — 48,675 
Balance at March 31, 2023106,869 $199 $1,260,310 $(332)$(813,405)$18,334 $465,106 
Stock-based compensation— — 19,947 — — — 19,947 
Exercise of stock options198 — 1,136 — — — 1,136 
Common stock - restricted stock units vesting35 — — — — —  
Foreign currency translation adjustments— — — (79)— — (79)
Interest in joint venture— — — — — (1,179)(1,179)
Net loss for the three months ended June 30, 2023— — — — (102,946)— (102,946)
Balance at June 30, 2023107,102 $199 $1,281,393 $(411)$(916,351)$17,155 $381,985 
Common
Stock
Amount ($)
Additional
Paid-In
Capital
Accumulated Other
Comprehensive
Income (Loss)
Accumulated
Deficit
Noncontrollling InterestTotals
Balance at March 31, 2021104,020 $196 $996,645 $102 $(551,394)$ $445,549 
Stock-based compensation— — 18,549 — — — 18,549 
Exercise of stock options161 2,755 — — — 2,756 
Common stock - restricted stock units vesting28 — — — — — — 
Foreign currency translation adjustments— — — (40)— — (40)
Net income (loss) for the three months ended June 30, 2021— — — — (29,924)— (29,924)
Balance at June 30, 2021104,209 $197 $1,017,949 $62 $(581,318)$ $436,890 
Common
Stock
Amount ($)
Additional
Paid-In
Capital
Accumulated Other
Comprehensive
Income (Loss)
Accumulated
Deficit
Noncontrollling InterestTotals
Balance at March 31, 2022105,702 $198 $1,115,373 $(107)$(663,198)$ $452,266 
Stock-based compensation— — 33,391 — — — 33,391 
Exercise of stock options53 — 599 — — — 599 
Common stock - restricted stock units vesting40 — — — — — — 
Foreign currency translation adjustments— — — (33)— — (33)
Interest in joint venture— — 39,750 — — 20,250 60,000 
Net income (loss) for the three months ended June 30, 2022— — — — (72,046)— (72,046)
Balance at June 30, 2022105,795 $198 $1,189,113 $(140)$(735,244)$20,250 $474,177 
Common
Stock
Amount ($)
Additional
Paid-In
Capital
Accumulated Other
Comprehensive
Income (Loss)
Accumulated
Deficit
Noncontrollling InterestTotals
Balance at September 30, 2020102,376 $195 $965,410 $18 $(503,844)$ $461,779 
Stock-based compensation— — 42,051 — — — 42,051 
Exercise of stock options981 10,489 — — — 10,490 
Common stock - restricted stock units vesting852 (1)— — — — 
Foreign currency translation adjustments— — — 44 — — 44 
Net income (loss) for the nine months ended June 30, 2021— — — — (77,474)— (77,474)
Balance at June 30, 2021104,209 $197 $1,017,949 $62 $(581,318)$ $436,890 
Common
Stock
Amount ($)
Additional
Paid-In
Capital
Accumulated Other
Comprehensive
Income (Loss)
Accumulated
Deficit
Noncontrollling InterestTotals
Common
Stock
Amount ($)
Additional
Paid-In
Capital
Accumulated
Other
Comprehensive
Loss
Accumulated
Deficit
Non-
controlling Interest
Totals
Balance at September 30, 2021Balance at September 30, 2021104,327 $197 $1,053,386 $(69)$(644,692)$ $408,822 Balance at September 30, 2021104,327 $197 $1,053,386 $(69)$(644,692)$ $408,822 
Stock-based compensationStock-based compensation— — 91,697 — — — 91,697 Stock-based compensation— — 24,504 — — — 24,504 
Exercise of stock optionsExercise of stock options497 — 4,281 — — — 4,281 Exercise of stock options208 — 2,145 — — — 2,145 
Common stock - restricted stock units vestingCommon stock - restricted stock units vesting971 (1)— — — — Common stock - restricted stock units vesting263 — — — — —  
Foreign currency translation adjustmentsForeign currency translation adjustments— — — (71)— — (71)Foreign currency translation adjustments— — — (39)— — (39)
Net loss for the three months ended December 31, 2021Net loss for the three months ended December 31, 2021— — — — (62,872)— (62,872)
Balance at December 31, 2021Balance at December 31, 2021104,798 $197 $1,080,035 $(108)$(707,564)$ $372,560 
Stock-based compensationStock-based compensation— — 33,802 — — — 33,802 
Exercise of stock optionsExercise of stock options237 — 1,537 — — — 1,537 
Common stock - restricted stock units vestingCommon stock - restricted stock units vesting667 (1)— — —  
Foreign currency translation adjustmentsForeign currency translation adjustments— — — — — 1 
Interest in joint ventureInterest in joint venture— — 39,750 — — 20,250 60,000 Interest in joint venture— — — — — —  
Net income (loss) for the nine months ended June 30, 2022— — — — (90,552)— (90,552)
Net income for the three months ended March 31, 2022Net income for the three months ended March 31, 2022— — — — 44,366 — 44,366 
Balance at March 31, 2022Balance at March 31, 2022105,702 $198 $1,115,373 $(107)$(663,198)$ $452,266 
Stock-based compensationStock-based compensation— — 33,391 — — — 33,391 
Exercise of stock optionsExercise of stock options53 — 599 — — — 599 
Common stock - restricted stock units vestingCommon stock - restricted stock units vesting40 — — — — —  
Foreign currency translation adjustmentsForeign currency translation adjustments— — — (33)— — (33)
Interest in joint ventureInterest in joint venture— — 39,750 — — 20,250 60,000 
Net loss for the three months ended June 30, 2022Net loss for the three months ended June 30, 2022— — — — (72,046)— (72,046)
Balance at June 30, 2022Balance at June 30, 2022105,795 $198 $1,189,113 $(140)$(735,244)$20,250 $474,177 Balance at June 30, 2022105,795 $198 $1,189,113 $(140)$(735,244)$20,250 $474,177 
The accompanying notes are an integral part of these unaudited consolidated financial statements.
3


Arrowhead Pharmaceuticals, Inc.
Consolidated Statements of Cash Flows
(in thousands)
(unaudited)
(In thousands, except per share amounts)
Nine Months Ended June 30,Nine Months Ended June 30,
2022202120232022
CASH FLOWS FROM OPERATING ACTIVITIES:CASH FLOWS FROM OPERATING ACTIVITIES:CASH FLOWS FROM OPERATING ACTIVITIES:
Net income (loss)$(90,552)$(77,474)
Net lossNet loss$(98,260)$(90,552)
Adjustments to reconcile net loss to net cash flow from operating activitiesAdjustments to reconcile net loss to net cash flow from operating activities
Stock-based compensationStock-based compensation91,697 42,051 Stock-based compensation59,949 91,697 
Depreciation and amortizationDepreciation and amortization7,761 5,763 Depreciation and amortization8,634 7,761 
Unrealized (gains) losses on marketable securities5,755 (1,387)
Amortization/(accretion) of note premiums/discounts2,013 142 
(Accretion) amortization of note premiums/discounts(Accretion) amortization of note premiums/discounts(1,030)2,013 
Non-cash interest expense on liability related to the sale of future royaltiesNon-cash interest expense on liability related to the sale of future royalties13,064 — 
Unrealized losses on marketable securitiesUnrealized losses on marketable securities— 5,755 
Changes in operating assets and liabilities:Changes in operating assets and liabilities:Changes in operating assets and liabilities:
Accounts receivableAccounts receivable10,016 174 Accounts receivable164 10,016 
Prepaid expenses and other current assetsPrepaid expenses and other current assets(8,876)(2,895)Prepaid expenses and other current assets27,913 (8,867)
Deferred revenue(87,100)211,392 
Accounts payableAccounts payable(3,563)3,208 Accounts payable5,001 (3,563)
Accrued expensesAccrued expenses1,713 13,682 Accrued expenses(32,082)1,713 
Other3,672 685 
NET CASH PROVIDED BY (USED IN) OPERATING ACTIVITIES(67,464)195,341 
Deferred revenueDeferred revenue(113,144)(87,100)
Operating lease liabilitiesOperating lease liabilities1,158 3,733 
Net cash used in operating activitiesNet cash used in operating activities(128,633)(67,394)
CASH FLOWS FROM INVESTING ACTIVITIES:CASH FLOWS FROM INVESTING ACTIVITIES:CASH FLOWS FROM INVESTING ACTIVITIES:
Purchases of property and equipmentPurchases of property and equipment(20,066)(15,368)Purchases of property and equipment(112,830)(20,066)
Purchases of investmentsPurchases of investments(223,391)(95,195)Purchases of investments(233,984)(223,391)
Proceeds from sale of investments201,595 87,130 
NET CASH PROVIDED BY (USED IN) INVESTING ACTIVITIES(41,862)(23,433)
Proceeds from maturities of investmentsProceeds from maturities of investments220,150 201,595 
Net cash used in investing activitiesNet cash used in investing activities(126,664)(41,862)
CASH FLOWS FROM FINANCING ACTIVITIES:CASH FLOWS FROM FINANCING ACTIVITIES:CASH FLOWS FROM FINANCING ACTIVITIES:
Proceeds from the exercises of stock optionsProceeds from the exercises of stock options4,331 10,490 Proceeds from the exercises of stock options2,232 4,331 
Proceeds from the sale of future royaltiesProceeds from the sale of future royalties250,000 — 
Proceeds from investment in joint ventureProceeds from investment in joint venture60,000 — Proceeds from investment in joint venture— 60,000 
NET CASH PROVIDED BY FINANCING ACTIVITIES64,331 10,490 
NET INCREASE (DECREASE) IN CASH(44,995)182,398 
CASH AND CASH EQUIVALENTS AT BEGINNING OF PERIOD184,434 143,583 
CASH AND CASH EQUIVALENTS AT END OF PERIOD$139,439 $325,981 
Proceeds from additional tenant improvement allowanceProceeds from additional tenant improvement allowance669 — 
Net cash provided by financing activitiesNet cash provided by financing activities252,901 64,331 
Net decrease in cash, cash equivalents and restricted cashNet decrease in cash, cash equivalents and restricted cash(2,396)(44,925)
Effect of exchange rate on cash, cash equivalents and restricted cashEffect of exchange rate on cash, cash equivalents and restricted cash(275)(70)
CASH, CASH EQUIVALENTS AND RESTRICTED CASH:CASH, CASH EQUIVALENTS AND RESTRICTED CASH:
BEGINNING OF PERIODBEGINNING OF PERIOD108,005 184,434 
END OF PERIODEND OF PERIOD$105,334 $139,439 
Supplementary disclosures:Supplementary disclosures:
Interest paidInterest paid$— $— 
Income taxes (paid) refundedIncome taxes (paid) refunded$— $— 
The accompanying notes are an integral part of these unaudited consolidated financial statements.
4


Arrowhead Pharmaceuticals, Inc.
Notes to Consolidated Financial Statements
(unaudited)
Unless otherwise noted, (1) the term “Arrowhead” refers to Arrowhead Pharmaceuticals, Inc., a Delaware corporation and its Subsidiaries, (2) the terms “Company,” “we,” “us,” and “our,” refer to the ongoing business operations of Arrowhead and its Subsidiaries, whether conducted through Arrowhead or a subsidiary of Arrowhead, (3) the term “Subsidiaries” refers to Arrowhead Madison Inc. (“Arrowhead Madison”), Visirna Therapeutics Inc. ("Visirna"), and Arrowhead Australia Pty Ltd (“Arrowhead Australia”), (4) the term “Common Stock” refers to Arrowhead’s Common Stock, par value $0.001 per share, (5) the term “Preferred Stock” refers to Arrowhead’s Preferred Stock, par value $0.001 per share, and (6) the term “Stockholder(s)” refers to the holders of Arrowhead’s Common Stock.
NOTE 1. ORGANIZATION AND SIGNIFICANT ACCOUNTING POLICIES
Nature of BusinessGeneral and Recent Developments
Arrowhead Pharmaceuticals, Inc. developsand its subsidiaries (referred to herein collectively as the “Company”) are primarily engaged in developing medicines that treat intractable diseases by silencing the genes that cause them. Using a broad portfolio of RNA chemistries and efficient modes of delivery, Arrowheadthe Company’s therapies trigger the RNA interference mechanism to induce rapid, deep and durable knockdown of target genes. RNA interference (“RNAi”) is a mechanism present in living cells that inhibits the expression of a specific gene, thereby affecting the production of a specific protein. Arrowhead’sThe Company’s RNAi-based therapeutics may leverage this natural pathway of gene silencing. silencing to target and shut down specific disease-causing genes.
The following table presents the Company’s pipeline includes ARO-APOC3 for hypertriglyceridemia, ARO-ANG3 for dyslipidemia, ARO-ENaC2 for cystic fibrosis, ARO-DUX4 for facioscapulohumeral muscular dystrophy, ARO-COV for the coronavirus that causes COVID-19 and other possible future pulmonary-borne pathogens, ARO-C3 for complement mediated diseases, ARO-RAGE and ARO-MUC5AC for various muco-obstructive or inflammatory pulmonary conditions and ARO-MMP7 for idiopathic pulmonary fibrosis. ARO-HSD for liver disease was out-licensed to Glaxosmithkline Intellectual Property (No. 3) Limited (“GSK”) in November 2021. ARO-XDH is being developed for uncontrolled gout under a collaboration agreement with Horizon Therapeutics Ireland DAC (“Horizon”). JNJ-75220795 (ARO-JNJ1) is being developed by Janssen as a potential treatment for patients with non-alcoholic steatohepatitis (NASH).current pipeline:ARO-AAT for liver disease associated with alpha-1 antitrypsin deficiency (“AATD”) was out-licensed to Takeda Pharmaceuticals U.S.A., Inc. (“Takeda”) in October 2020. JNJ-3989 (formerly referred to as ARO-HBV) for chronic hepatitis B virus was out-licensed to Janssen in October 2018. Olpasiran (formerly referred to as AMG 890 or ARO-LPA) for cardiovascular disease was out-licensed to Amgen Inc. (“Amgen”) in 2016. While the Company believes that initial ARO-HIF2 Phase 1 clinical data provides proof of concept for the ability to deliver siRNA to RCC tumors, the Company has decided not to pursue further clinical development of ARO-HIF2 based on a number of factors including the evolving competitive landscape for HIF2 inhibitors.
Therapeutic AreaNameStageProduct Rights
CardiometabolicARO-APOC3Phase 2b and Phase 3Arrowhead
ARO-ANG3Phase 2bArrowhead
OlpasiranPhase 3Amgen
PulmonaryARO-ENAC2Pre-ClinicalArrowhead
ARO-RAGEPhase 1/2Arrowhead
ARO-MUC5ACPhase 1/2aArrowhead
ARO-MMP7Phase 1/2aArrowhead
LiverGSK-4532990 (formerly ARO-HSD)Phase 2GSK
FazirsiranPhase 3Takeda and Arrowhead
JNJ-3989Phase 2Janssen
HZN-457 (formerly ARO-XDH)Phase 1Horizon
ARO-C3Phase 1/2Arrowhead
ARO-PNPLA3 (formerly JNJ-75220795)Phase 1Arrowhead
MuscleARO-DUX4Pre-ClinicalArrowhead
Central Nervous System (CNS)ARO-SOD1Pre-ClinicalArrowhead
The Company operates lab facilities in Madison, Wisconsin and San Diego, California and Madison, Wisconsin, where the Company’sits research and development activities, including the development of RNAi therapeutics, take place. The Company’s principal executive offices are located in Pasadena, California.
During the first three quarters of fiscal 2022,2023, the Company continued to develop and advance its pipeline and partnered candidates and expanded its facilities to support the Company’s growing pipeline.candidates. Several key recent developments include:
i)dosedhosted a Research & Development (R&D) Day on June 1, 2023 to discuss progress of the first patients in its PALISADE study, a phase 3 clinical study to evaluateCompany's pipeline of RNAi Therapeutics, at which the safetyfollowing updates were discussed:
ARO-RAGE showed continued dose response with single inhaled dose of 184 mg achieving mean knockdown of 90% and efficacymax of ARO-APOC3 in adults with familial chylomicronemia syndrome (FCS)95%;
ii)entered into an exclusive license agreementadipose delivery platform achieved single dose target gene silencing of greater than 90% with GSK for ARO-HSD;six months of duration in non-human primates;
iii)Janssen presented clinical data from REEF-1, a Phase 2b studyimproved hepatic dimer platform achieved equivalent or better knockdown of different combination regimens, including JNJ-73763989 (JNJ-3989), formerly called ARO-HBV, and/or JNJ-56136379 (JNJ-6379), and a nucleos(t)ide analog (NA) for the treatment of chronic hepatitis B virus infection (CHB);two target genes with longer duration than monomer mixture in non-human primates;
iv)filed for regulatory clearanceTRiM™ platform now has potential to begin a Phase 1/2a study of ARO-C3address multiple cell types including liver, solid tumors, lung, central nervous system, skeletal muscle, and subsequently dosed the first subjects in AROC3-1001, a Phase 1/2 clinical study of ARO-C3, the Company’s investigational RNA interference (RNAi) therapeutic designed to reduce production of complement component 3 (C3) as a potential therapy for various complement mediated diseases;adipose;
v)presented additional interim clinical data from AROHSD1001, AROAAT2002, and AROAPOC31001;announced progress towards the Company's "20 in 25" goal to grow its pipeline of RNAi therapeutics that leverage the proprietary Targeted RNAi Molecule (TRiM™) platform to a total of
5


vi)completed the purchase of 13 acres of land20 clinical stage or marketed products in the Verona Technology Park in Verona, Wisconsin and held a groundbreaking ceremony on the site. The site is being developed into an approximately 160,000 square foot drug manufacturing facility and an approximately 140,000 square foot laboratory and office facility which will support the Company's process development and analytical activities. The Company also announced that it received awards for up to $16 million in tax increment financingyear 2025;
presented updated data from the city of Verona, and up to $2.5 million in refundable Wisconsin state income tax credits from the Wisconsin Economic Development Corporation (WEDC) as incentives to invest in the local community and create new jobs. Additionally, The Company entered into a lease agreement for a new 144,000 square foot laboratory and office facility in San Diego, California to support discovery activities;
vii)completed enrollment in Phase 2b ARCHES-22 SEQUOIA study of investigational ARO-ANG3 forRNAi therapy Fazirsiran in patients with mixed dyslipidemia;alpha-1 antitrypsin deficiency liver disease which included:
viii)filed for regulatory clearance to initiate Phase 1/2a study of ARO-RAGE for treatment of Asthma;Fazirsiran reduced serum Z-AAT concentration in a dose-dependent manner;
ix)filed for regulatory clearance to initiate Phase 1/2a study of ARO-MUC5AC for treatment of muco-obstructive lung disease;Fazirsiran significantly reduced liver Z-AAT;
x)initiated and dosedFazirsiran consistently reduced hepatic globule burden;
Fazirsiran treatment reduced histological signs of hepatic inflammation;
50% of the firstpooled Fazirsiran treated patients showed at least a one-point improvement in METAVIR liver fibrosis versus 38% in the placebo group;
Fazirsiran has been well tolerated to date;
pulmonary function test results (FEV1 and DLCO) for both Fazirsiran and placebo were stable over time with no apparent dose-dependent effects;
updated Phase 2 clinical data were presented at the European Association for the Study of the Liver (EASL) Congress 2023 in an oral presentation titled, “Fazirsiran reduces liver Z-alpha-1 antitrypsin synthesis, decreases globule burden and improves histological measures of liver disease in adults with alpha-1 antitrypsin deficiency: a randomized placebo-controlled phase 2 study”;
presented interim data from the ongoing Phase 2 GATEWAY clinical study of ARO-ANG3 which included:
mean reduction in LDL-C of 48.1% (200mg) and 44.0% (300mg);
ANPTL3 inhibition with ARO-ANG3 also reduced HDL-C, non-HDL-C, and triglycerides, consistent with published human genetic data;
safety and tolerability;
completed enrollment of the Phase 3 PALISADE clinical trial evaluating ARO-APOC3 for treatment of familial chylomicronemia syndrome;
announced interim results from ARO-RAGE administration in Part 1 of the ongoing Phase 1/2 study in normal healthy volunteers which included:
reductions in soluble RAGE (sRAGE) as measured in serum after two doses on Day 1 and Day 29;
duration of pharmacologic effect persisted for at least 6 weeks after the second administration of the 92 mg does with further follow up ongoing;
reduction in sRAGE as measured in bronchoalveolar lavage fluid (BALF) at Day 31 after a single dose;
reduction in in serum sRAGE was observed after a single dose;
the pooled placebo groups experienced a mean sRAGE increase of 8% in BALF and a mean decrease of 1% serum;
safety and tolerability;
expanded TRiMTM platform to include an optimized intrathecal administration for CNS delivery with distribution throughout the brain and in all relevant brain cell types. The first development candidate to utilize this new delivery platform is ARO-SOD1. In June 2023, the Company filed a clinical trial application (CTA) for approval to initiate a Phase 1 clinical study. In preclinical studies, ARO-SOD1 achieved 95% spinal cord tissue mRNA knockdown after a single intrathecal dose in human SOD1 transgenic rats and maintained greater than 80% spinal cord tissue mRNA knockdown three months after a single intrathecal dose in non-human primates;
dosed the first patient in Takeda’s Phase 3 REDWOOD clinical study of Fazirsiran for the treatment of alpha-1 antitrypsin deficiency associated liver diseases, triggering a $40.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023;
dosed the first patient in GSK’s Phase 2b trial of GSK4532990, formerly called ARO-HSD, an investigational ARO-ANG3RNAi therapeutic for the treatment of patients with homozygousnon-alcoholic steatohepatitis (NASH), triggering a $30.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023;
6


announced that the U.S. Food and Drug Administration (FDA) has granted Fast Track designation to ARO-APOC3 for reducing triglycerides in adult patients with familial hypercholersterolemia;chylomicronemia syndrome (FCS). ARO-APOC3 was previously granted Orphan Drug designation by the FDA and the European Union;
xi)entered into definitive agreementsannounced interim results from Part 1 of AROC3-1001, an ongoing Phase 1/2 clinical study of ARO-C3, which included:
a dose-dependent reduction in serum C3, with 88% mean reduction at highest dose tested;
a dose-dependent reduction in AH50, a marker of alternative complement pathway hemolytic activity, with 91% mean reduction at highest dose tested;
duration of pharmacologic effect supportive of quarterly or less frequent subcutaneous dose administration;
safety and tolerability;
received notice from Janssen of its decision to form a joint venture, Visirna Therapeutics, Inc. (“Visirna”voluntarily terminate the Research Collaboration and Option Agreement (the “Janssen Collaboration Agreement”) with Vivo Capital (“Vivo”) through whichbetween the Company and Vivo intendJanssen. The Company regained full rights to expandARO-PNPLA3, formerly called JNJ-75220795, upon termination of the reach of innovative medicinesJanssen Collaboration Agreement, which took effect on April 7, 2023. ARO-PNPLA3 is in Greater China;Phase 1 clinical trials that are now being developed by the Company;
xii)hostedinitiated dosing in ARO-MMP7-1001 (NCT05537025), a pulmonary research & development (R&D) DayPhase 1/2a single ascending dose and multiple ascending dose clinical study to discussevaluate the Company’s emerging pipelinesafety, tolerability, pharmacokinetics, and pharmacodynamics of pulmonary targeted RNA interference (RNAi)ARO-MMP7, an investigational RNAi therapeutic candidates that leverage its proprietary Targeted RNAi Molecule (TRIMTM) platform, including an announcement of its previously undisclosed candidate designed to reduce expression of matrix metalloproteinase 7 (MMP7) as a potential treatment for idiopathic pulmonary fibrosis (IPF);, in up to 56 healthy volunteers and in up to 21 patients with IPF;
xiii)enrolled the first subject in conjunctiona Phase 1 randomized, placebo-controlled trial to assess the safety tolerability, pharmacokinetics and pharmacodynamics of a development-stage medicine, HZN-457 (previously known as ARO-XDH), which is out-licensed to Horizon, triggering a $15.0 million milestone payment to the Company which was paid in the second quarter of fiscal 2023;
enrolled the first subject in Amgen’s Phase 3 trial of Olpasiran, triggering a $25.0 million milestone payment to the Company which was paid in the second quarter of fiscal 2023;
entered into a Royalty Purchase Agreement (the “Royalty Pharma Agreement”) with Takeda, Royalty Pharma Investments 2019 ICAV (“Royalty Pharma”) on November 9, 2022, pursuant to which Royalty Pharma paid $250.0 million upfront (See Note 11 — Liability Related to the Sale of Future Royalties of Notes to Consolidated Financial Statements of Part I, “Item 1. Financial Statements.”);
announced top line results from athe SEQUOIA Phase 2 clinicalStudy of Fazirsiran in patients with Alpha-1 Antitrypsin Deficiency-Associated Liver Disease in which:
fibrosis regression was observed in 50% of patients receiving Fazirsiran;
median reductions of 94% of Z-AAT accumulation in the liver and mean reductions of 68% in histologic globule burden were observed;
treatment emergent adverse events were generally well balanced between Fazirsiran and placebo groups;
results were consistent with AROAAT-2002 open-label study (AROAAT-2002) of investigational fazirsiran (TAK-999/ARO-AAT) for the treatment of liver disease associated with alpha-1 antitrypsin deficiency (AATD), and was recentlypreviously published in theThe New England Journal of Medicine (NEJM)Medicine.
Consolidation and presentedBasis of Presentation
The interim Consolidated Financial Statements include the accounts of Arrowhead Pharmaceuticals, Inc. and its subsidiaries (wholly-owned subsidiaries and a variable interest entity for which the Company is the primary beneficiary). Subsidiaries refer to Arrowhead Madison, Inc., Visirna Therapeutics, Inc. (“Visirna”), and Arrowhead Australia Pty Ltd. For subsidiaries in an oral presentation at The International Liver Congress™ 2022 - The Annual Meetingwhich the Company owns or is exposed to less than 100% of the European Association foreconomics, the StudyCompany records net loss attributable to noncontrolling interests in its consolidated statements of operations equal to the percentage of the Liver (EASL).economic or ownership interests retained in such entity by the respective noncontrolling party.
The Company is actively monitoring the ongoing COVID-19 pandemic. The financial results for the three and nine months ended June 30, 2022 were not significantly impacted by COVID-19. Operationally, the Company has experienced delays in its earlier stage programs due to a shortage in non-human primates, which are critical to the Company’s preclinical programs. Additionally, the Company has experienced delays in enrollment in its clinical trials. The Company’s operations at its research and development facilities in Madison, Wisconsin and San Diego, California, and its corporate headquarters in Pasadena, California have continued with limited impact, other than for enhanced safety measures and intermittent lab supply shortages. However, the Company cannot predict the impact the progression of COVID-19 will have on future financial and operational results due to a variety of factors, including the ability of the Company’s clinical sites to continue to enroll subjects, the ability of the Company’s suppliers to continue to operate, the continued good health and safety of the Company’s employees and the length and severity of the COVID-19 pandemic.
Liquidity
Theinterim Consolidated Financial Statements have been prepared in conformity with theU.S. generally accepted accounting principles generally accepted in the United States of America (“GAAP”), which contemplate the continuation. The financial data of the Company asincluded herein are unaudited. In the opinion of management, all material adjustments of a going concern. Historically,normal recurring nature have been made to present fairly the Company’s
7


financial position at June 30, 2023 and the results of operations and cash flows for the periods presented. All intercompany transactions and balances have been eliminated. Certain prior period amounts have been reclassified to conform with the current period presentation.
Certain financial information that is normally included in annual financial statements prepared in accordance with GAAP, but that is not required for interim reporting purposes, has been omitted from the accompanying interim consolidated financial statements and related notes. Readers are urged to review the Company’s Annual Report on Form 10-K for the year ended September 30, 2022 for more complete descriptions and discussions. Operating results and cash flows for the nine months ended June 30, 2023 are not necessarily indicative of the results that may be expected for the fiscal year ending September 30, 2023.
Liquidity
The Company’s primary sources of financing have been through the sale of its securities, and revenue from its licensing and collaboration agreements.agreements and the sale of certain future royalties. Research and development activities have required significant capital investment since the Company’s inception and are expected to continue to require significant cash expenditure in the future, particularly as the Company’s pipeline of drug candidates and its headcount have both expanded significantly. Additionally, significant capital investment will be required as the Company’s pipeline matures into later stage clinical trials and as well as with the Company’sCompany plans to increase its internal manufacturing capabilities, and expand its footprint in Verona, Wisconsin and San Diego, California.
6


capabilities.
At June 30, 2022,2023, the Company had $139.4$105.3 million in cash and cash equivalents (including $7.4$7.3 million in restricted cash), $277.1$346.4 million in short-term investments $0 in marketable securities and $165.9$42.8 million in long-term investments to fund operations. $60 million of our cash balance resulted from the formation of our joint venture, Viserna. During the nine months ended June 30, 2022,2023, the Company’s cash and cash equivalents and investments balance decreasedincreased by $31.0$12.2 million which was primarily due to cash being used to fund the Company’s operations, partially offset by the $120.0$250.0 million upfront payment received from GSK,Royalty Pharma (Note 11) and $60$110.0 million in milestone payments from the Company’s collaboration and license agreements, partially offset by cash infusionused to Viserna.fund its operations.
In total, the Company remainsis eligible for $4.9to receive up to $3.4 billion in developmental, regulatory and sales milestones, and may receive various royalties on net sales from its licensing and collaboration agreements, subject to the terms and conditions of those agreements. The revenue recognition for these collaboration agreements is discussed further in Note 2 below.2.
Summary of Significant Accounting Policies
There have been no changes to the significant accounting policies disclosed in the Company’s most recent Annual Report on Form 10-K.10-K for the fiscal year ended September 30, 2022.
Recent Accounting Pronouncements
There have been no recent accounting pronouncements that have significantly impacted this Quarterly Report on Form 10-Q, beyond those disclosed in the Company’s most recent Annual Report on Form 10-K.

Basis of Presentation

The accompanying unaudited consolidated financial statements have been prepared in accordance with accounting principles generally accepted in the United States of America (“US GAAP”) for interim financial information. Accordingly, they do not include all of the information and notes required by US GAAP for complete financial statements. In our opinion, the unaudited consolidated financial statements have been prepared on the same basis as audited consolidated financial statements and include all adjustments, consisting of only normal recurring adjustments, necessary10-K for the fair presentation of our financial position, results of operations, comprehensive income and cash flows. The interim results are not necessarily indicative of the results of operations to be expected for the year ending September 30, 2022 or any other period.

The accompanying unaudited consolidated financial statements include the accounts of our wholly owned subsidiaries, as well as the accounts of Visirna, a variable interest entity for which we are the primary beneficiary. All intercompany balances and transactions have been eliminated in consolidation. For consolidated entities where we own or are exposed to less than 100% of the economics, we record net income (loss) attributable to noncontrolling interest in our unaudited consolidated statements of income equal to the percentage of the economic or ownership interest retained in such entities by the respective noncontrolling parties. The accompanying unaudited consolidated financial statements should be read in conjunction with the audited consolidated financial statements and notes thereto included in our Annual Report on Form 10-K for thefiscal year ended September 30, 2021, filed with the SEC.2022.
NOTE 2. COLLABORATION AND LICENSE AGREEMENTS
Amgen Inc.The following table provides a summary of revenue recognized:
On September 28, 2016,
Three Months Ended June 30,Nine Months Ended June 30,
2023202220232022
(in thousands)
GSK$277 $— $29,600 $120,000 
Horizon1,539 6,666 23,206 21,251 
Takeda14,009 25,507 146,477 67,100 
Janssen— 239 355 3,305 
Amgen— — 25,000 — 
Total$15,825 $32,412 $224,638 $211,656 
The following table summarizes the Company entered into 2 collaborationbalance of receivables and license agreements and a common stock purchase agreement with Amgen. Under the Second Collaboration and License Agreement (the “Olpasiran Agreement”), Amgen has received a worldwide, exclusive licensecontract liabilities related to Arrowhead’s novel RNAi Olpasiran (previously referred to as AMG 890 or ARO-LPA) program. These RNAi molecules are designed to reduce elevated lipoprotein(a), which is a genetically validated, independent risk factor for atherosclerotic cardiovascular disease. Under the prior collaboration and license agreement (the “First Collaboration and License Agreement” or the “ARO-AMG1 Agreement”), Amgen received an option to a worldwide, exclusive license for ARO-AMG1, an RNAi therapy for an undisclosed genetically validated cardiovascular target. Under both agreements, Amgen is wholly responsible for clinical development and commercialization. Under the terms of the agreements taken together, the Company has received $35.0 million in upfront payments, $21.5 million in the form of an equity investment by Amgen in the Company’s Common Stock, and $30.0 million in milestone payments, and may receive up to an additional $400.0 million in remaining development, regulatory and sales milestone payments. The Company is further eligible to receive up to low double-digit royalties for sales of products under the Olpasiran Agreement. In July 2019, Amgen informed the Company that it would not be exercising its option for an exclusive license for ARO-AMG1, and as such, there will be no further milestone or royalty payments under
78


the ARO-AMG1 Agreement.collaboration and license agreements:
The Company has evaluated these agreements in accordance with FASB Topics 808 – Collaboration Arrangements and 606 - Revenue for Contracts from Customers. The Company has substantially completed its performance obligations under the Olpasiran Agreement and the ARO-AMG1 Agreement. Future milestones and royalties achieved will be recognized in their entirety when earned. In July 2020, Amgen initiated a Phase 2 clinical study of Olpasiran, which resulted in a $20.0 million milestone payment to the Company. During the three and nine months ended June 30, 2022 and 2021, the Company recognized $0 and $0 of revenue associated with its agreement with Amgen, respectively.As of June 30, 2022, there were $0 in contract assets recorded as accounts receivable and $0 contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.
June 30, 2023September 30, 2022
(in thousands)
Receivables included in accounts receivable$1,539 $6,174 
Contract liabilities included in deferred revenue$16,905 $130,049 
Janssen Pharmaceuticals, Inc.Glaxosmithkline Intellectual Property (No. 3) Limited (“GSK”)
On October 3, 2018,November 22, 2021, GSK and the Company entered into aan Exclusive License Agreement (the “Janssen“GSK License Agreement”) and a Research Collaboration and Option Agreement (the “Janssen Collaboration Agreement”) with Janssen, part of the Janssen Pharmaceutical Companies of Johnson & Johnson. The Company also entered into a stock purchase agreement with JJDC (“JJDC Stock Purchase Agreement”). Under the JanssenGSK License Agreement, JanssenGSK has received a worldwide,an exclusive license tofor GSK-4532990 (formerly ARO-HSD). The exclusive license is worldwide with the Company’s JNJ-3989 (ARO-HBV) program, the Company’s third-generation subcutaneously administered RNAi therapeutic candidate being developed as a potential therapy for patients with chronic hepatitis B virus infection. Beyond the Company’sexception of greater China. The Company completed its Phase 1/2 study of JNJ-3989 (ARO-HBV), which the Company was responsible for completing, JanssenGSK-4532990, and GSK is wholly responsible for all clinical development and commercialization of JNJ-3989. Under the Janssen Collaboration Agreement, Janssen was able to select three new targets against which Arrowhead would develop clinical candidates. These candidates were subject to certain restrictions and do not include candidates that already wereGSK-4532990 in the Company’s pipeline. The Company was obligated to perform discovery, optimization and preclinical research and development, entirely funded by Janssen, which on its own or in combination with Janssen development work, would have been sufficient to allow the filing of a U.S. Investigational New Drug Application or equivalent, at which time Janssen would have the option to take an exclusive license. If the option was exercised, Janssen would have been wholly responsible for clinical development and commercialization of each optioned candidate.territory. Under the terms of the agreements taken together,agreement, the Company has received $175.0 million as an upfront payment $75.0 million in the form of an equity investment by JJDC in Arrowhead Common Stock under the JJDC Stock Purchase Agreement, and milestone and option payments totaling $73.0$120.0 million and recognized an additional $30.0 million at the start of a Phase 2 trial. The Company is also eligible for an additional payment of $100.0 million upon achieving a successful Phase 2 trial readout and the first patient dosed in a Phase 3 trial. Furthermore, should the Phase 3 trial read out positively, and the potential new medicine receives regulatory approval in major markets, the deal provides for commercial milestone payments to the Company may receiveof up to $1.6 billion in development and sales milestones payments for the Janssen License Agreement,$190.0 million at first commercial sale, and up to $0.6 billion$590.0 million in development and salessales-related milestone payments for the remaining target covered under the Janssen Collaboration Agreement.payments. The Company is further eligible to receive tiered royalties on net product sales upin a range of mid-teens to mid-teens under the Janssen License Agreement and up to low teens under the Janssen Collaboration Agreement. During the three months ended June 30, 2022, Janssen’s option period expired unexercised for two of the three candidates (ARO-JNJ2 and ARO-JNJ3) under the Janssen Collaboration Agreement.twenty percent.
The Company has evaluated these agreements in accordance with FASB Topics 808 – Collaboration Arrangements and 606 - Revenue for Contracts from Customers. At the inception of these agreements,the GSK License Agreement, the Company identified 1one distinct performance obligation. Regarding the Janssen License Agreement, theThe Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibility to complete the Phase 1/2 study of JNJ-3989 (ARO-HBV) and the Company’s responsibility to ensure certain manufacturing of JNJ-3989 (ARO-HBV) drug product is completed and delivered to Janssen (the “Janssen“GSK R&D Services”). Due to the specialized and unique nature of these Janssenthe GSK R&D Services and their direct relationship with the license, the Company determined that these deliverables representrepresented one distinct bundle and, thus, one performance obligation. The Company also determined that Janssen’s option to requireBeyond the GSK R&D Services, which are the responsibility of the Company, to develop up to three new targets is not a material rightGSK will be responsible for managing future clinical development and thus, not a performance obligation at the onset of the agreement. The consideration for this option is accounted for separately.commercialization in its territory.
The Company determined the initial transaction price totaled approximately $252.7$120.0 million, which includesincluding the upfront payment, the premium paid by JJDC for its equity investmentwhich was collected in theJanuary 2022. The Company two $25.0 million milestone payments relatedhas excluded any future estimated milestones or royalties from this transaction price to JNJ-3989 (ARO-HBV), and estimated payments for reimbursable Janssen R&D Services to be performed.date. The Company has allocated the total $252.7$120.0 million initial transaction price to its one distinct performance obligation for the JNJ-3989 (ARO-HBV)GSK-4532990 license and the associated JanssenGSK R&D Services. As the Company has completed its performance obligation related to this agreement, the upfront payment of $120.0 million was fully recognized during the six months ended March 31, 2022. Further, GSK dosed the first patient in a Phase 2 trial in March 2023, triggering a $30.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023. There were no contract assets and liabilities recorded as of June 30, 2023.
Horizon Therapeutics Ireland DAC (“Horizon”)
On June 18, 2021, Horizon and the Company entered into a collaboration and license agreement (the “Horizon License Agreement”). Under the terms of the Horizon License Agreement, Horizon received a worldwide exclusive license for HZN-457, a clinical-stage medicine being developed by Horizon as a potential treatment for people with uncontrolled gout. The Company conducted all activities through the preclinical stages of development of, and Horizon is now wholly responsible for clinical development and commercialization of, HZN-457. The Company received $40.0 million as an upfront payment in July 2021 and an additional $15.0 million upon Horizon’s initiation of a Phase 1 clinical trial in January 2023, and is eligible to receive up to $645.0 million in additional potential development, regulatory and sales milestones. The Company is also eligible to receive royalties in the low- to mid-teens range on net product sales.
At the inception of the Horizon License Agreement, the Company identified one distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibilities to conduct all activities through the preclinical stages of development of HZN-457 (the “Horizon R&D Services”). Due to the specialized and unique nature of these Horizon R&D Services and their direct relationship with the license, the Company determined that these deliverables represented one distinct bundle and, thus, one performance obligation. Beyond the Horizon R&D Services, which are the responsibility of the Company, Horizon is responsible for managing future clinical development and commercialization of HZN-457.
The Company determined the initial transaction price totaled $40.0 million, including the upfront payment. The Company has recognizedexcluded any future estimated milestones or royalties from this transaction price into date. The Company allocated the total $40.0 million initial transaction price to its entirety as of September 30, 2021, as its performance obligations were substantially completed. Future milestones and royalties achieved will be recognized in their entirety when earned. During the three months ended June 30, 2022 and 2021, the Company recognized approximately $0 and $0 of revenue associated with thisone distinct performance obligation respectively. Duringfor the nine months ended June 30, 2022HZN-457 license and 2021, the Companyassociated Horizon R&D Services. Revenue was recognized approximately $0 and $20.2 million of revenue associated with this performance obligation, respectively. As of June 30, 2022, there were $0 in contracton a straight-line basis over the timeframe for
89


assets recorded as accounts receivable, and $0 of contract liabilities recorded as current deferred revenue oncompleting the Company’s Consolidated Balance Sheets.
Horizon R&D Services. The Company has conducteddetermined that the straight-line basis was appropriate as its discovery, optimizationefforts were expended evenly over the course of completing its performance obligation. Further, Horizon enrolled the first subject in December 2022 in a Phase 1 randomized, placebo-controlled trial to assess the safety, tolerability, pharmacokinetics and preclinical research and developmentpharmacodynamics of JNJ-75220795 (ARO-JNJ1), ARO-JNJ2, and ARO-JNJ3 under the Janssen Collaboration Agreement. All costs and labor hours spent byHZN-457, triggering a $15.0 million milestone payment to the Company have been entirely funded by Janssen. During the three months ended June 30, 2022, Janssen’s option period expired unexercised for two of the three candidates (ARO-JNJ2 and ARO-JNJ3) under the Janssen Collaboration Agreement as reportedwhich was paid in the second quarter of 2022. During the three months ended June 30, 2022 and 2021, the Company recognized $0.0fiscal 2023. There was $1.5 million and $0.2 million of revenue associated with these efforts, respectively. During the nine months ended June 30, 2022 and 2021, the Company recognized $0.1 million and $0.5 million of revenue associated with these efforts, respectively. In May 2021, Janssen exercised its option right for ARO-JNJ1, which resulted in a $10.0 million milestone payment to the Company. This $10 million milestone payment was recognized entirely during the three months ended June 30, 2021.As of June 30, 2022, there were $0 of contract assets recorded as accounts receivable and $0 of contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.of June 30, 2023.
Takeda Pharmaceuticals U.S.A., Inc.Pharmaceutical Company Limited (“Takeda”)
On October 7, 2020, Takeda and the Company entered into an Exclusive License and Co-funding agreementCo-Funding Agreement (the “Takeda License Agreement”) with Takeda.. Under the Takeda License Agreement, Takeda and the Company will co-develop the Company’s ARO-AATits Fazirsiran program, the Company’s second-generation subcutaneously administered RNAi therapeutic candidate being developed as a treatment for liver disease associated with alpha-1 antitrypsin deficiency. Within the United States, ARO-AAT,Fazirsiran, if approved, will be co-commercialized under a 50/50 profit sharing structure. Outside the United States, Takeda will lead the global commercialization strategy and will receivereceived an exclusive license to commercialize ARO-AAT,Fazirsiran, while the Company will be eligible to receive tiered royalties of 20% to 25% on net sales. In January 2021, theThe Company received $300.0 million as an upfront payment in January 2021, recognized an additional $40.0 million upon Takeda’s initiation of a Phase 3 clinical study in March 2023, and is eligible to receive potential development, regulatory and commercial milestones of up to $740.0$527.5 million.
The Company has evaluated the Takeda License Agreement in accordance with FASB Topics 808 – Collaborative Arrangements and 606 - Revenue for Contracts from Customers. At the inception of the Takeda License Agreement, the Company identified 1one distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services including the Company’s responsibilities to complete the initial portion of the SEQUOIA study, to complete the ongoing Phase 2 AROAAT2002 study and to ensure certain manufacturing of ARO-AATFazirsiran drug product is completed and delivered to Takeda (the “Takeda R&D Services”). Due to the specialized and unique nature of these Takeda R&D Services and their direct relationship with the license, the Company determined that these deliverables represent 1one distinct bundle and, thus, one performance obligation. Beyond the Takeda R&D Services, which are the responsibility of the Company, Takeda will be responsible for managing future clinical development and commercialization outside the United States. Within the United States, the Company will also participate in co-development and co-commercialization efforts and will co-fund these efforts with Takeda as part of the 50/50 profit sharing structure within the United States. The Company considers the collaborative activities, including the co-development and co-commercialization, to be a separate unit of account within Topic 808, and as such, these co-funding amounts will beare recorded as Researchresearch and Development Expensesdevelopment expenses or Generalgeneral and Administrative Expenses,administrative expenses, as appropriate.
The Company determined the initial transaction price totaled $300.0 million, which includes the upfront payment. The Company has excluded any future milestones or royalties from this transaction price to date. The Company has allocated the total $300.0 million initial transaction price to its one distinct performance obligation for the ARO-AATFazirsiran license and the associated Takeda R&D Services. Revenue will beis recognized using a proportional performance method (based on actual patient visits completed versus total estimated visits completed for the ongoing SEQUOIA and AROAAT2002 clinical studies). RevenueThe Company previously expected these clinical trials to extend to September 2025 in order to demonstrate long term safety and efficacy in the open label extension (OLE) part of the studies; however, Takeda now intends to initiate a new OLE study available to patients participating in these Phase 2 studies that will initiate as early as the fourth quarter of fiscal 2023. Based on this new information, patients enrolled in the SEQUOIA and AROAAT2002 studies are expected to complete their Phase 2 study visits between June 2023 and June 2024, shortening the Company’s performance obligation. As a result, effective the second quarter of fiscal 2023, the Company changed its estimates of the revenue recognition to better reflect this newly estimated performance period. The effect of these changes in estimates resulted in accelerated revenue by $61.4 million, or $0.58 per share (diluted) for each of the three months ended June 30, 2022 and 2021 was $25.5 million and $35.7 million, respectively. Revenue for the nine months ended June 30, 2022 and 2021 was $67.12023. There were $16.9 million and $69.3 million, respectively. As of June 30, 2022, there were $0 in contract assets recorded as accounts receivable, $71.0 million in contract liabilities recorded as deferred revenue, and $71.2of which $16.9 million was classified as current as of June 30, 2023.
In March 2023, Takeda dosed the first patient in contract liabilitiesthe Phase 3 REDWOOD clinical study of Fazirsiran, triggering a $40.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023. The Company also recorded as deferred revenue, net of the current portion, and $7.8$1.4 million in contract liabilities recorded as accrued expenses. The $7.8 million in accrued expenses as of June 30, 2023 that was primarily driven by co-development and co-commercialization activities.
Horizon Therapeutics Ireland DACJanssen Pharmaceuticals, Inc. (“Janssen”)
On June 18, 2021,October 3, 2018, Janssen, part of the Janssen Pharmaceutical Companies of Johnson & Johnson, and the Company entered into the Horizona License Agreement (the “Janssen License Agreement”) and the Janssen Collaboration Agreement. The Company also entered into a stock purchase agreement with Horizon. JJDC, Inc. (“JJDC”), Johnson & Johnson’s venture capital arm (the “JJDC Stock Purchase Agreement”).
Under the HorizonJanssen License Agreement, Janssen received a worldwide, exclusive license to the Company’s JNJ-3989 (ARO-HBV) program, the Company’s third-generation subcutaneously administered RNAi therapeutic candidate being
910


License Agreement, Horizon received a worldwide exclusive license for ARO-XDH, a previously undisclosed discovery-stage investigational RNAi therapeutic being developed by the Company as a potential treatment for people with uncontrolled gout. The Company will conduct all activities through the preclinical stages of development of ARO-XDH, and Horizon will be wholly responsible for clinical development and commercialization of ARO-XDH. In July 2021, the Company received $40 million as an upfront payment and is eligible to receive up to $660 million in potential development, regulatory and sales milestones. The Company is also eligible to receive royalties in the low- to mid-teens range on net product sales.
The Company has evaluated the Horizon License Agreement in accordance with FASB Topics 808 – Collaborative Arrangements and 606 - Revenue for Contracts from Customers. At the inception of the Horizon License Agreement, the Company identified one distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibilities to conduct all activities through the preclinical stages of development of ARO-XDH (the “Horizon R&D Services”). Due to the specialized and unique nature of these Horizon R&D Services and their direct relationship with the license, the Company determined that these deliverables represented 1 distinct bundle and, thus, 1 performance obligation. Beyond the Horizon R&D Services, which are the responsibility of the Company, Horizon will be responsible for managing future clinical development and commercialization of ARO-XDH.
The Company determined the initial transaction price totaled $40.0 million, including the upfront payment. The Company has excluded any future estimated milestones or royalties, from this transaction price to date. The Company will allocate the total $40.0 million initial transaction price to its 1 distinct performance obligation for the ARO-XDH license and the associated Horizon R&D Services. Revenue will be recognized on a straight-line basis over the estimated timeframe for completing the Horizon R&D Services. The Company determined that the straight-line basis was appropriate as its efforts will be expended evenly over the course of completing its performance obligation. Revenue for the three months ended June 30, 2022 and 2021 was $6.7 million and $0, respectively. Revenue for the nine months ended June 30, 2022 and 2021 was $20.0 million and $0, respectively. As of June 30, 2022, there were $0 million in contract assets recorded as accounts receivable, $13.3 million in contract liabilities recorded as deferred revenue.
The Company has manufactured ARO-XDH material for Horizon in furtherance of the research plan entered into pursuant to the Horizon License Agreement, for which the Company has been reimbursed for its costs. During the nine months ended June 30, 2022 and 2021, the Company recognized $1.3 million and $0 with these efforts, respectively. As of June 30, 2022, there were $0.0 million of contract assets recorded as accounts receivable and $0 of contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.
Glaxosmithkline Intellectual Property (No. 3) Limited
On November 22, 2021, the Company entered into an Exclusive License Agreement (the “GSK License Agreement”) with GSK. Under the GSK License Agreement, GSK has received an exclusive license for ARO-HSD, the Company’s investigational RNAi therapeutic being developed as a treatmenttherapy for patients with alcohol-related and nonalcohol related liver diseases, such as nonalcoholic steatohepatitis (NASH). The exclusive license is worldwide with the exception of greater China, for which the Company retained rights to develop and commercialize. chronic hepatitis B virus infection. Beyond the Company’s Phase 1/2 study of (ARO-HSD)JNJ-3989 (ARO-HBV), which the Company iswas responsible for completing, GSKJanssen is wholly responsible for clinical development and commercialization of ARO-HSD in its territory.JNJ-3989 (ARO-HBV). Under the terms of the agreement,Janssen License Agreement, the Company has received $175.0 million as an upfront payment, $75.0 million in the form of $120an equity investment by JJDC in the Company’s common stock under the JJDC Stock Purchase Agreement, and milestone and option payments totaling $73.0 million, and is eligible for additional payments of $30 million at the start of Phase 2Company may receive up to $0.8 billion in development and $100 million upon achieving a successful Phase 2 trial readout and the first patient dosed in a Phase 3 trial. Furthermore, should the Phase 3 trial read out positively, and the potential new medicine receives regulatory approval in major markets, the deal provides for commercialsales milestone payments tofor the Company of up to $190 million at first commercial sale, and up to $590 million in sales-related milestone payments.Janssen License Agreement. The Company is further eligible to receive tiered royalties on net product sales up to mid-teens under the Janssen License Agreement.
In May 2021, Janssen exercised its option right for JNJ-75220795 (ARO-JNJ1) which resulted in a range$10.0 million milestone payment to the Company. This $10.0 million milestone payment was recognized entirely as of mid-teens to twenty percent.
September 30, 2021. The Company has evaluatedconducted its discovery, optimization and preclinical research and development of JNJ-75220795 (ARO-JNJ1), ARO-JNJ2, and ARO-JNJ3 under the GSK License Agreement in accordance with FASB Topics 808 – Collaborative ArrangementsJanssen Collaboration Agreement. All costs and 606 - Revenuelabor hours spent by the Company have been entirely funded by Janssen. On April 7, 2023, Janssen voluntarily terminated the Janssen Collaboration Agreement. Upon termination, the Company regained full rights to ARO-PNPLA3, formerly called JNJ-75220795, the only candidate for Contracts from Customers. which Janssen had exercised its option.
At the inception of the GSKJanssen License Agreement, the Company identified 1 distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services including the Company’s responsibility to complete the Phase 1/2 study of JNJ-3989 (ARO-HBV) and the Company’s responsibility to ensure certain manufacturing of JNJ-3989 (ARO-HBV) drug product is completed and delivered to Janssen (the “GSK“Janssen R&D Services”). Due to the specialized and unique nature of these GSKJanssen R&D Services and their direct relationship with the license, the Company determined that these deliverables represented 1represent one distinct bundle and, thus, one performance obligation. Beyond the GSK R&D Services, which are the responsibility of the Company, GSK will be responsible for managing future clinical development and commercialization in its territory.
10


The Company determined the initial transaction price totaled $120.0approximately $252.7 million, includingwhich includes the upfront payment. The $120.0payment, the premium paid by JJDC for its equity investment in the Company, two $25.0 million upfront payment was collected in January 2022. The Company has excluded any futuremilestone payments related to JNJ-3989 (ARO-HBV), and estimated milestones or royalties from this transaction pricepayments for reimbursable Janssen R&D Services to date.be performed. The Company has allocated the total $120.0$252.7 million initial transaction price to its one distinct performance obligation for the ARO-HSDJNJ-3989 (ARO-HBV) license and the associated GSKJanssen R&D Services. AsThe Company recognized this transaction price in its entirety as of September 30, 2021, as its performance obligations were substantially completed. Future milestones and royalties achieved will be recognized in their entirety when earned. There were no contract assets and liabilities recorded as of June 30, 2023.
Amgen Inc. (“Amgen”)
On September 28, 2016, Amgen and the Company entered into two collaboration and license agreements and a common stock purchase agreement. Under the Second Collaboration and License Agreement (the “Olpasiran Agreement”), Amgen received a worldwide, exclusive license to the Company’s novel RNAi Olpasiran program. These RNAi molecules are designed to reduce elevated lipoprotein(a), which is a genetically validated, independent risk factor for atherosclerotic cardiovascular disease. Under the first collaboration and license agreement (the “First Collaboration and License Agreement” or the “ARO-AMG1 Agreement”), Amgen received an option to a worldwide, exclusive license to ARO-AMG1, an RNAi therapy for an undisclosed genetically validated cardiovascular target. Under both agreements, Amgen is wholly responsible for clinical development and commercialization.
Under the Olpasiran Agreement and the ARO-AMG1 Agreement, the Company has received $35.0 million in upfront payments and $21.5 million in the form of an equity investment by Amgen in the Company’s common stock. Further, the Company received additional an $55.0 million in milestone payments; $10.0 million upon Amgen’s initiation of Phase 1 study in September 2018,$20.0 million upon its initiation of a Phase 2 clinical study in July 2020, and $25.0 million upon its first subject enrollment in a Phase 3 trial in December 2022. The Company has substantially completed its performance obligation related to this agreement,obligations under the upfront payment of $120.0 million will be fully recognizedOlpasiran Agreement and the ARO-AMG1 Agreement. There were no contract assets and liabilities recorded as of the three and nine months ended June 30, 2022. Revenue for the nine months ended June 30, 2022 and 2021 was $120.0 million and $0, respectively. As of June 30, 2023.
In November 2022, there were $0Royalty Pharma and the Company entered into the Royalty Pharma Agreement. In consideration for the payments under the Royalty Pharma Agreement, Royalty Pharma is entitled to receive all royalties otherwise payable by Amgen to the Company under the Olpasiran Agreement. The Company remains eligible to receive up to an additional $535.0 million in contract assets recorded as accounts receivable, $0 in contract liabilities recorded as deferred revenue.

remaining development, regulatory and sales milestone payments payable from Amgen and Royalty Pharma. See Note 11.
Joint Venture and License Agreement with Visirna Therapeutics, Inc. (“Visirna”)

On April 25, 2022, Visirna and the Company entered into a License Agreement (the “Visirna License Agreement”) with Visirna Therapeutics, Inc. (“Visirna”), pursuant to which Visirna received an exclusive license to develop, manufacture and commercialize four of Arrowhead’s the Company’s
11


RNAi-based investigational cardioboliccardiometabolic medicines in Greater China (including the People’s Republic of China, Hong Kong, Macau and Taiwan).Pursuant to a Share Purchase Agreement (the “Visirna SPA”) entered into simultaneously with the Visirna License Agreement, (the “Visirna SPA”), the Company acquired a majority stake in Visirna (after accounting for shares reserved for Visirna’s employee stock ownership plan) as partial consideration for the Visirna License Agreement.Under the Visirna SPA, entities affiliated with Vivo Capital (“Vivo”) also acquired a minority stake in Visirna in exchange for $60$60.0 million in upfront capital to support the operations of Visirna.As further consideration under the Visirna License Agreement, the Company is also eligible to receive potential royalties on commercial sales.

During the nine months ended June 30, 2023, the Company performed manufacturing and development work pursuant to a Clinical Supply Agreement between the parties contemplated by the Visirna License Agreement. The Company has determined that Visirna is a variable interest entity upon its formationreceived $0.9 million as consideration for this manufacturing and development work, and there were no contract assets and liabilities recorded as of June 30, 2022, and it has been determined that Arrowhead is the primary beneficiary; accordingly, Arrowhead consolidates the financial statements of Visirna.2023.
12


NOTE 3. PROPERTY AND EQUIPMENT
The following table summarizes the Company’s major classes of property and equipment:
June 30, 2022September 30, 2021June 30, 2023September 30, 2022
(In thousands)(in thousands)
Computers, software, office equipment and furnitureComputers, software, office equipment and furniture$2,210 $2,170 Computers, software, office equipment and furniture$2,198 $2,182 
LandLand2,996 2,996 
Research equipmentResearch equipment31,678 27,500 Research equipment50,472 38,283 
Leasehold improvementsLeasehold improvements42,017 41,524 Leasehold improvements96,344 42,017 
Construction in Progress22,338 345 
Land2,996 — 
Total gross fixed assets101,239 71,539 
Construction in progressConstruction in progress118,279 56,373 
270,289 141,851 
Less: Accumulated depreciation and amortizationLess: Accumulated depreciation and amortization(29,335)(22,864)Less: Accumulated depreciation and amortization(38,920)(31,554)
Property and equipment, netProperty and equipment, net$71,904 $48,675 Property and equipment, net$231,369 $110,297 
Depreciation and amortization expense for property and equipment for the three months ended June 30, 2023 and 2022 and 2021 was $2.2$2.9 million and $1.6$2.2 million, respectively. Depreciation and amortization expense for property and equipment for the nine months ended June 30, 2023 and 2022 was $7.4 million and 2021 was $6.5 million, and $4.5 million, respectively. Construction
The increase in Progress relatesthe construction in progress during the nine months ended June 30, 2023 was mainly due to the Company’scontinuing developments of manufacturing, laboratory and office facilities in Verona, Wisconsin as well as a new laboratory and office facility in San Diego, research facilities. Land relates toCalifornia. In May 2023, the Company’s Verona, Wisconsin research facility.
NOTE 4. INVESTMENTSCompany completed the development of the San Diego facility, which resulted in the reclassification of construction in progress as leasehold improvements as of June 30, 2023. See Note 7.

1113


Investments at June 30, 2022 primarilyNOTE 4. INVESTMENTS
The Company’s investments consisted of commercial paper and corporate bonds that have maturities of less than 36 months and a certificate of deposit. Cash and cash equivalents consist of cash on hand and commercial paper purchased with 90-days or less remaining to maturity. The Company’s corporate bonds consist of both short-term and long-term bonds and are classified as “held-to-maturity” on the Company’s Consolidated Balance Sheets. The Company’s certificate of deposit matures in less than 12 months and is classified as “held-to-maturity” on the Company’s Consolidated Balance Sheet,’. The Company’s marketable equity securities consisted of mutual funds that primarily invest in U.S. government bonds, U.S. government agency bonds, corporate bonds and other asset-backed debt securities. Dividends from these funds were automatically re-invested. In April 2022, all marketable securities were sold for $122.3 million. The Company may also invest excess cash balances in money market accounts, and these securities would be classified as cash and cash equivalents on the Company’s Consolidated Balance Sheet. The Company accounts for its “held-to-maturity” investments in accordance with FASB ASC 320, Investments – Debt and Equity Securities and its marketable equity securities in accordance with ASC 321, Investments – Equity Securities. We did not record any impairment charges related to our marketable debt securities during the three and nine months ended June 30, 2022.following:
The following tables summarize the Company’s short-term and long-term investments and marketable securities as of June 30, 2022 and September 30, 2021 by measurement category:
As of June 30, 2023
(in thousands)
Adjusted BasisGross
Unrealized Gains
Gross
Unrealized Losses
Fair Value
Short-term investments (due within one year)
Held to maturity debt securities$346,369 $— $(4,204)$342,165 
Held to maturity certificate of deposit— — — — 
Total short-term investments$346,369 $— $(4,204)$342,165 
Long-term investments (due within one through three years)
Held to maturity debt securities$42,758 $— $(294)$42,464 
Total long-term investments$42,758 $— $(294)$42,464 
As of June 30, 2022
(In thousands)
Cost BasisGross
Unrealized
Gains
Gross
Unrealized
Losses
Fair Value
Classified as Cash Equivalents    
Commercial notes (due within ninety days)$25,325 $11 $— $25,336 
Classified as Held to Maturity    
Commercial notes (due within one year)$227,057 $21 $(1,588)$225,490 
Commercial notes (due within one through three years)$165,920 $— $(6,380)$159,540 
Certificate of deposit (due within one year)$50,000 $— $— $50,000 
Classified as Marketable Securities    
Marketable securities$— $— $— $— 
Total$468,302 $32 $(7,968)$460,366 
As of September 30, 2021
(In thousands)
Cost BasisGross
Unrealized
Gains
Gross
Unrealized
Losses
Fair Value
Classified as Held to Maturity    
Commercial notes (due within one year)$56,627 $803 $— $57,430 
Commercial notes (due within one through three years)$195,595 $1,151 $(103)$196,643 
Certificate of deposit (due within one through two years)$50,000 $— $— $50,000 
Classified as Marketable Securities    
Marketable securities$127,481 $— $(753)$126,728 
Total$429,703 $1,954 $(856)$430,801 
As of September 30, 2022
(in thousands)
Adjusted BasisGross
Unrealized Gains
Gross
Unrealized Losses
Fair Value
Short-term investments (due within one year)
Held to maturity debt securities$218,391 $— $(3,661)$214,730 
Held to maturity certificate of deposit50,000 — — 50,000 
Total short-term investments$268,391 $— $(3,661)$264,730 
Long-term investments (due within one through three years)
Held to maturity debt securities$105,872 $— $(5,569)$100,303 
Total long-term investments$105,872 $— $(5,569)$100,303 
14


NOTE 5. INTANGIBLE ASSETS
Intangible assets subject to amortization include patents and a license agreement capitalized as part of the Novartis RNAi asset acquisition in March 2015. The license agreement associatedfollowing table presents the components of intangible assets:
Gross Carrying AmountAccumulated AmortizationImpairmentNet Carrying AmountUseful Lives
(in thousands)(in years)
As of June 30, 2023
Patents$21,728 $12,933 $— $8,795 14
License3,129 1,237 — 1,892 21
Total intangible assets, net$24,857 $14,170 $— $10,687 
As of September 30, 2022
Patents$21,728 $11,770 $— $9,958 14
License3,129 1,125 — 2,004 21
Total intangible assets, net$24,857 $12,895 $— $11,962 
Intangible assets are reviewed annually for impairment and more frequently if potential impairment indicators exist. No impairment indicators were identified during the nine months ended June 30, 2023 and 2022.
Intangible assets with the Novartis RNAi asset acquisition is beingdefinite useful lives are amortized on a straight-line basis over the estimated life remaining at the timetheir useful lives. Intangible assets amortization expense was $0.4 million for each of acquisition, which was 21 years, and the accumulated amortization of the asset is $1.1 million. The patents associated with the Novartis RNAi asset acquisition are being amortized over the estimated life remaining at the time of acquisition, which was 14 years, and the accumulated
12


amortization of the assets is $11.4 million. Amortization expense for the three months ended June 30, 2023 and 2022, and 2021 was $0.4$1.3 million and $0.4 million, respectively. Amortization expense for each of the nine months ended June 30, 20222023 and 2021 was $1.3 million and $1.3 million, respectively. Amortization expense is expected2022. None of the intangible assets with definite useful lives are anticipated to be $0.4 million for the remainder of fiscal2022, $1.7 million in 2023, $1.7 million in 2024, $1.7 million in 2025, $1.7 million in 2026 and $5.2 million thereafter.have a residual value.
The following table provides details onpresents the Company’sestimated future amortization expense related to intangible asset balances:assets as of June 30, 2023:
Intangible
Assets
Subject to
Amortization
(in thousands)
Balance at September 30, 2021$13,663 
Impairment— 
Amortization(1,276)
Balance at June 30, 2022$12,387 
Amortization Expense
Year Ending September 30,(in thousands)
2023 (remainder)$425 
20241,700 
20251,700 
20261,700 
20271,700 
Thereafter3,462 
Total$10,687 

15


NOTE 6. STOCKHOLDERS’ EQUITY
AtThe following table summarizes the Company’s shares of common stock and preferred stock:
Shares
Par ValueAuthorizedIssuedOutstanding
(in thousands)
As of June 30, 2023
Common stock$0.001 290,000 107,102 107,102 
Preferred stock$0.001 5,000 — — 
As of September 30, 2022
Common stock$0.001 145,000 105,960 105,960 
Preferred stock$0.001 5,000 — — 
On March 16, 2023, the Company’s stockholders approved an increase in authorized common shares, par value 0.001 per share, from 145,000,000 to 290,000,000. The amendment to the Amended and Restated Certificate of Incorporation was filed on April 27, 2023.
As of June 30, 2023 and September 30, 2022, the Company had a total of 150,000,000respectively, 12,914,571 and 14,000,392 shares of capitalcommon stock authorized for issuance, consisting of 145,000,000 shares of Common Stock, par value $0.001 per share, and 5,000,000 shares of Preferred Stock, par value $0.001 per share.
At June 30, 2022, 105,795,456 shares of Common Stock were outstanding. At June 30, 2022, 13,942,716 shares of Common Stock were reserved for issuance upon exercise of options and vesting of restricted stock units granted or available for grant under Arrowhead’sthe Company’s 2004 Equity Incentive Plan, 2013 Incentive Plan, and 2021 Incentive Plan, as well as for inducement grants made to new employees under Rule 5635(c)(4) of the Nasdaq Listing Rules.
In August 2020,On December 2, 2022, the Company entered into an Openopen market sale agreement (the “Open Market Sale Agreement (the “ATM Agreement”), pursuant to which the Company may, from time to time, sell up to $250,000,000 in shares of the Company’s Common Stockcommon stock through Jefferies LLC, acting as the sales agent and/or principal, in an at-the-market offering.offering (“ATM Offering”). The Company is not required to sell shares under the ATMOpen Market Sale Agreement. The Company will pay Jefferies LLC a commission of up to 3.0% of the aggregate gross proceeds received from all sales of the common stock under the ATMOpen Market Sale Agreement. Unless otherwise terminated, the ATM Agreement continues untilOffering shall terminate upon the earlier of selling(i) the sale of all shares available underof common stock subject to the ATMSales Agreement or December 2, 2022. Atand (ii) the termination of the Sales Agreement as permitted therein. The Company and Jefferies may each terminate the Open Market Sale Agreement at any time upon prior notice. As of June 30, 2022,2023, no shares have been soldissued under the ATMOpen Market Sale Agreement.
NOTE 7. COMMITMENTS AND CONTINGENCIES
Litigation
From time to time, the Company may be subject to various claims and legal proceedings in the ordinary course of business. If the potential loss from any claim, asserted or unasserted, or legal proceeding is considered probable and the amount is reasonably estimable, the Company will accrue a liability for the estimated loss. There were no contingent liabilities recorded as of June 30, 2022.2023.
Commitments
On December 20, 2021, the Company completed a purchase of 13 acres of land in the Verona Technology Park in Verona, Wisconsin, which is planned to be the site ofbeing developed into an approximately 160,000 square foot drug manufacturing facility and an approximately 140,000 square foot laboratory and office facility towhich will support the Company’s process development and analytical activities. ArrowheadAs of June 30, 2023, the Company has incurred $102.7 million and intends to invest between $200spend an additional $160.0 million and $250to $180.0 million intoto complete the buildoutbuild out of the facilities. As part of this acquisition, the Company also entered into a development agreement with the City of Verona to construct certain infrastructure improvements within the TIFtax incremental district and will be reimbursed up to $16.0 million by the City of Verona by future tax increment revenue generated from the developed property. The total amount of funding that the City of Verona will pay as reimbursements under the TIFTax Incremental Financing program for these improvements is not guaranteed and will depend on future tax revenues generated from the developed property.
13


The Company will also receive up to $2.5 million of refundable Wisconsin state income tax credits from the Wisconsin Economic Development Corporation (WEDC) as incentives to invest in the local community and create new jobs.
Technology License Commitments
The Company has licensed from third parties the rights to use certain technologies for its research and development activities, as well as in any products the Companyit may develop using these licensed technologies. These agreements and other similar
16


agreements often require the Company to make milestone and royalty payments. Milestone payments, for example, may be required as the research and development process progresses through various stages of development, such as when clinical candidates enter or progress through clinical trials, upon a new drug application and uponNDA and/or certain sales level milestones. These milestone payments could amount to the mid to upper double-digit millions of dollars. During the three and nine months ended June 30, 20222023 and 2021,2022, the Company did not reach any milestones. Under certain agreements, the Company may be required to make mid to high single-digit percentage royalty payments based on a percentage of sales of the relevant products.
NOTE 8. LEASES
Leases
In April 2019, the Company entered into a lease for its corporate headquarters in Pasadena, California. The 91 month office building lease between the Company and 177 Colorado Owner, LLC is for approximately 24,000 square feet of office space located at 177 E. Colorado Blvd, Pasadena, California. The increased capacity of this new office space compared to the Company’s prior corporate headquarters will accommodate increased personnel as the Company’s pipeline of drug candidates expands and moves closer to market. Lease payments began on September 30, 2019 and are estimated to total approximately $8.7 million over the term. The lease expires on April 30, 2027. The Company has paid approximately $3.5 million for leasehold improvements, net of tenant improvement allowances. The lease contains an option to renew for 1 term of five years. The exercise of this option was not determined to be reasonably certain and thus was not included in lease liabilities on the Company’s Consolidated Balance Sheet at June 30, 2022. On October 23, 2020, the Company entered into a lease expansion to add an additional approximately 24,000 square feet of office space at the same location for its corporate headquarters. Lease payments for the expansion began in July 2021and the lease for the expansion expires in April 2027. The lease payments for the expansion are expected to total $6.9 million. The Company has paid approximately $4.0 million of leasehold improvements, net of tenant improvement allowances, for the lease expansion.
In January 2016, the Company entered into a lease for its research facility in Madison, Wisconsin. The lease was for approximately 60,000 square feet of office and laboratory space and had an expiration date of September 30, 2026. The lease was amended in January 2019 and May 2020 to expand the rentable square feet by an additional 40,000 square feet and to extend the lease expiration date to September 30, 2031. Lease payments are estimated to total approximately $26.2 million for the term.The Company incurred approximately $11.0 million of leasehold improvements for the additional 40,000 square feet, net of tenant improvement allowances. The lease contains 2 options to renew for two terms of five years. The exercise of these options were not determined to be reasonably certain and thus was not included in lease liabilities on the Company’s Consolidated Balance Sheet at June 30, 2022. In November 2020 and December 2020, the Company entered into amendments to expand the rentable square space by an additional 10,743 square feet and these amendments added a total of approximately $1.2 million of lease payments for the remainder of the term.
In March 2020, the Company entered into a sublease agreement for additional research and development facility space in San Diego, California. The Sublease provides additional space needed to accommodate the recent growth of the Company’s personnel and discovery efforts. The Sublease is for approximately 21,000 rentable square feet. The term of the Sublease commenced on April 1, 2020 and will end on January 14, 2023. Sublease payments are estimated to total approximately $2.0 million over the term.
On November 19, 2021, the Company entered into a new lease for a San Diego, California research facility. The 15-year lease is for approximately 144,000 square feet of office and research and development laboratory space to be constructed in San Diego, California. This lease will replace the Company’s current research facility sublease for property located in San Diego, California. The increased capacity of this new facility compared to the Company’s current research facility in San Diego will accommodateaccommodates increased personnel for the Company’sits expanding pipeline of current and future drug candidates. The estimated rent commencement date for the lease is in March 2023, after construction and leasehold improvements have been completed. The lease payments, which beginbegan on April 19, 2023, the rent commencement date, will be approximately $119.0 million over the initial 15-year term. The Company also estimates payments forannual operating expenses to be approximately $3.0 million for the first year of the lease, and these payments will continue throughout the initial 15-year term. The Company expects to pay approximately $31.0$32.0 million for leasehold improvements, net of tenant improvement allowances. Pursuant to the lease, within twelve months of the expiration of the initial 15-year term, the
14


Company has the option to extend the lease for up to 1one additional ten-year term, with certain annual increases in base rent. .
OperatingFurther, the lease agreement grants the Company the right to receive an Additional Tenant Improvement Allowance (“ATIA”) funded by the lessor. The maximum amount of ATIA is $7.2 million, and as of June 30, 2023, the Company has received approximately $0.7 million, which has been recorded as other liabilities on its consolidated balance sheets. The Company will repay the ATIA through equal monthly payments, including 7% interest per annum over the base term, starting from the rent commencement date. Interest begins accruing on the date the lessor first disburses the ATIA.
Other Significant Leases
Pasadena, California: The Company leases 49,000 square feet of office space located at 177 Colorado Blvd. for its corporate headquarters from 177 Colorado Owner, LLC, which lease expires on April 30, 2027. The lease contains an option to renew for one term of five years.
San Diego, California: The Company subleased space from Halozyme, Inc. for additional research and development space in San Diego, California. The term of this sublease commenced on April 1, 2020 and ended on January 14, 2023. On December 23, 2022, the Company entered into a new six-month lease agreement with 11404 & 11408 Sorrento Valley Owner (DE) LLC, effective January 15, 2023. The lease ended on July 15, 2023.
Madison, Wisconsin: The Company leases space for office and laboratory facilities, which expires on September 30, 2031. The lease contains options to renew for two terms of five years. After accounting for additional rental square feet added pursuant to amendments to the lease agreement in 2019 and 2020, the Company currently leases a total of 111,000 square feet.
The components of lease assets and liabilities along with their classification on the Company’s consolidated balance sheets were as follows:
Lease Assets and LiabilitiesClassificationJune 30, 2023September 30, 2022
(in thousands)
Operating lease assetsRight-of-use assets$40,667 $58,291 
Current operating lease liabilitiesLease liabilities2,823 2,776 
Non-current operating lease liabilitiesLease liabilities, net of current portion79,911 78,800 
Three Months Ended June 30,Nine Months Ended June 30,
Lease CostClassification2023202220232022
(in thousands)
Operating lease costResearch and development$3,323 $2,974 $7,735 $4,757 
General and administrative expense509 448 1,542 1,288 
Variable lease cost (1)
Research and development257 179 627 519 
General and administrative expense— — — — 
Total$4,089 $3,601 $9,904 $6,564 
17


(1) Variable lease cost is primarily related to operating expenses associated with the Company’s operating leases.
There was $0.6 million and $0.2 million short-term lease cost during the three months ended June 30, 2023, and 2022, and 2021respectively. There was $3.4$1.2 million and $1.6$0.7 million respectively. Operatingshort-term lease cost during the nine months ended June 30, 2023, and 2022, and 2021 was $6.0 million and $3.6 million, respectively. Variable lease costs for the three months ended June 30, 2022 and 2021 was $0.2 million and $0.1 million, respectively. Variable lease costs for the nine months ended June 30, 2022 and 2021 was $0.5 million and $0.6 million, respectively. There was no short-term lease cost during the three and nine months ended June 30, 2022 and 2021.
The following table presents payments of operating lease liabilities on an undiscounted basis as of June 30, 2022:2023:
YearYearAmounts
(in thousands)(in thousands)
2022 (remainder of fiscal year)$1,309 
20235,802 
2023 (remainder of fiscal year)2023 (remainder of fiscal year)$1,143 
202420248,094 20248,094 
2025202511,800 202511,800 
2026202612,313 202612,148 
2027 and thereafter114,109 
2027202711,320 
2028 and thereafter2028 and thereafter102,812 
TotalTotal$153,427 Total$147,317 
Less imputed interestLess imputed interest$(72,313)Less imputed interest$(64,583)
Total operating lease liabilities (includes current portion)Total operating lease liabilities (includes current portion)$81,114 Total operating lease liabilities (includes current portion)$82,734 
Cash paid for the amounts included in the measurement of the operating lease liabilities on the Company’s Consolidated Balance SheetSupplemental cash flow and included in Other changes in operating assets and liabilities within cash flows from operating activities on the Company’s Consolidated Statements of Cash Flows for the nine months ended June 30, 2022 and 2021other information related to leases was $3.4 million and $2.2 million, respectively. The weighted-average remaining lease term and weighted-average discount rate for all leases as of June 30, 2022 was 7.3 years and 8.5%, respectively.follows:
Nine Months Ended June 30,
20232022
Cash paid for amounts included in the measurement of lease liabilities:
Operating cash flows from operating leases (in thousands)4,430 3,398 
June 30,
20232022
Weighted-average remaining lease term (in years)13.47.3
Weighted-average discount rate8.0 %8.5 %
18


NOTE 9. STOCK-BASED COMPENSATION
ArrowheadThe Company has three plans that provide for equity-based compensation. Under the 2004 Equity Incentive Plan (the “2004 Plan”) and the 2013 Incentive Plan (the “2013 Plan”), 68,555 and 3,440,076 shares, respectively, of the Company’s common stock are reserved for grants of stock options and restricted stock awards to employees and directors as of June 30, 2022, 216,607 and 4,114,9992023.
On March 18, 2021, the Company’s Board of Directors approved the Arrowhead Pharmaceuticals, Inc. 2021 Incentive Plan (the “2021 Plan”), which authorized 8,000,000 shares respectively, of Arrowhead’s Common Stock are reserved(subject to certain adjustments) available for the grantgrants of stock options, stock appreciation rights, restricted and restrictedunrestricted stock, unitperformance awards, cash awards and other awards convertible into or otherwise based on shares of the Company’s common stock. The maximum number of shares authorized under the 2021 Plan will be (i) reduced by any shares subject to employees, consultants and others. No further grants may beawards made under the 2004 Equity Incentive Plan.2013 Plan after January 1, 2021, and (ii) increased by any shares subject to outstanding awards under the 2013 Plan as of January 1, 2021 that, after January 1, 2021, are canceled, expired, forfeited or otherwise not issued under such awards (other than as a result of being tendered or withheld to pay the exercise price or withholding taxes in connection with any such awards) or settled in cash. As of June 30, 2022, there2023, the total number of shares reserved for issuance was 6,186,644 shares, which included 197,596 shares that were options granted and outstanding to purchase 216,607 and 1,806,987 shares of Common Stock under the 2004 Equity Incentive Plan and the 2013 Incentive Plan, respectively, and there were 2,308,012 restricted stock units granted and outstandingforfeited under the 2013 IncentivePlan, and 1,977,114 shares have been granted under the 2021 Plan. As of June 30, 2022,
In addition, there were 816,248712,454 shares reserved for options and 682,500746,175 shares reserved for restricted stock units issued as inducement grants to new employees granted outside of equitythe Company’s equity-based compensation plans. Asplans under Rule 5635(c)(4) of the Nasdaq Listing Rules.
The following table presents a summary of awards outstanding:
As of June 30, 2023
2004 Plan2013 Plan2021 PlanInducement AwardsTotal
Granted and outstanding awards:
Options68,555 1,550,951 33,838 712,454 2,365,798 
Restricted stock units— 1,889,125 1,686,766 746,175 4,322,066 
Total68,555 3,440,076 1,720,604 1,458,629 6,687,864 
The following table summarizes stock-based compensation expenses included in operating expenses:
Three Months Ended June 30,Nine Months Ended June 30,
2023202220232022
(in thousands)
Research and development8,982 8,098 26,129 23,958 
General and administrative10,965 25,292 33,820 67,739 
Total$19,947 $33,390 $59,949 $91,697 
Stock Option Awards
The following table presents a summary of the stock option activity for the nine months ended June 30, 2022, there were 3,000 shares2023:
SharesWeighted-
Average
Exercise
Price
Per Share
Weighted-
Average
Remaining
Contractual
Term (Years)
Aggregate
Intrinsic
Value
Outstanding at September 30, 20222,721,384$20.73 
Granted32,15133.03 
Cancelled or expired(43,936)62.31 
Exercised(343,801)6.48 
Outstanding at June 30, 20232,365,798$22.20 4.4 years$43,813,599 
Exercisable at June 30, 20232,188,690$20.28 4.2 years$43,480,339 
The aggregate intrinsic values represents the amount by which the market price of Common Stock reserved forthe underlying stock exceeds the exercise price of the option. The total intrinsic value of the options and 1,507,267 shares of Common Stock reserved for restricted stock units granted and outstanding underexercised during the 2021 Incentive Plan. As ofthree months ended June 30, 2022, the total number of shares available under the 2021 Incentive Plan was 6,627,845 shares, which includes 119,612 shares that were forfeited under the 2013 Incentive Plan.2023 and
1519


Stock Options
2022 was $6.5 million and $1.6 million, respectively. The following table summarizes information about stock options:
Number of
Options
Outstanding
Weighted-
Average
Exercise
Price
Per Share
Weighted-
Average
Remaining
Contractual
Term
Aggregate
Intrinsic
Value
Balance at September 30, 20213,456,239$19.60 
Granted— 
Cancelled(106,801)43.68 
Exercised(506,596)8.45 
Balance at June 30, 20222,842,842$20.60 5.1 years$55,434,252 
Exercisable at June 30, 20222,370,089$16.14 4.6 years$52,941,990 
total intrinsic value of the options exercised during the nine months ended June 30, 2023 and 2022 was $10.1 million and $24.9 million, respectively.
Stock-based compensation expense related to stock options outstanding for the three months ended June 30, 2023 and 2022, and 2021 was $2.6$2.1 million and $3.2$2.6 million, respectively. Stock-based compensation expense related to stock options for the nine months ended June 30, 2023 and 2022 and 2021 was $8.3$6.7 million and $9.6 million, respectively. For non-qualified stock options, the expense creates a timing difference, resulting in a deferred tax asset, which is fully reserved by a valuation allowance.
The grant date fair value of the options granted by the Company for the three months ended June 30, 2022 and 2021 was $0 and $0.9 million, respectively. The grant date fair value of the options granted by the Company for the nine months ended June 30, 2022 and 2021 was $0 and $9.0 million, respectively.
The intrinsic value of the options exercised during the three months ended June 30, 2022 and 2021 was $1.6 million and $10.2 million, respectively. The intrinsic value of the options exercised during the nine months ended June 30, 2022 and 2021 was $24.9 million and $63.0$8.3 million, respectively.
As of June 30, 2022,2023, the pre-tax compensation expense for all outstanding unvested stock options in the amount of $14.9$5.3 million will be recognized in the Company’s results of operations over a weighted average period of 1.7 years.12 months.
The fair value of each stock option award is estimated on the date of grant using the Black-Scholes option pricing model. The Black-Scholes option valuation model was developed for use in estimating the fair value of traded options, which do not have vesting restrictions and are fully transferable. The determination of the fair value of each stock option is affected by the Company’s stock price on the date of grant, as well as assumptions regarding a number of highly complex and subjective variables. Because the Company’s employee stock options have characteristics significantly different from those of traded options, and because changes in the subjective input assumptions can materially affect the fair value estimate, in management’s opinion, the existing models do not necessarily provide a reliable single measure of the fair value of its employee stock options.
The following table provides the assumptions used to valuein the calculation of grant-date fair values of these stock options are as follows:based on the Back-Scholes option pricing model:
Nine Months Ended June 30,
20222021
Dividend yieldN/A— 
Risk-free interest rateN/A0.4% - 1.1%
VolatilityN/A86% - 90.4%
Expected life (in years)N/A6.25
Weighted average grant date fair value per share of options grantedN/A$48.64 
Nine Months Ended June 30,
2023
2022 (5)
Expected dividend yield(1)
— N/A
Risk-free interest rate(2)
3.69 %N/A
Expected volatility(3)
86.4 %N/A
Expected term (in years)(4)
6.25N/A
Weighted average grant date fair value per share of options granted$24.80 N/A
(1) The dividend yield is zero as the Company currently does not pay a dividend.
(2) The risk-free interest rate is based on that of the U.S. Treasury bond.yields with equivalent terms in effect at the time of the grant.
(3) Volatility is estimated based on volatility average of the Company’s Common Stockcommon stock price.
(4) The expected term represents the period of time that stock options granted are expected to be outstanding, by using historical exercise patterns and post-vesting termination behavior.
16


(5) No options were granted during the nine months ended June 30, 2022.
Restricted Stock Units
Restricted stock units (“RSUs”), including market-based, time-based market condition-based, and performance condition-basedperformance-based awards, have been granted under the Company’s 2013 Incentive Plan,and 2021 Incentive Plan,Plans and as inducements grants granted outside of the Company’s equity-based compensation plans under Rule 5635(c)(4) of the Nasdaq Listing Rules.plans. At vesting, each outstanding RSU will be exchanged for one share of the Company’s Common Stock.common stock. RSU awards generally vest subject to the satisfaction of service requirements or the satisfaction of both service requirements and achievement of certain performance targets.
The following table summarizes the activity of the Company’s RSUs:
Number of
RSUs
Weighted-
Average
Grant
Date
Fair Value
Per Share
Number of
RSUs
Weighted-
Average
Grant
Date
Fair Value
Per Share
Unvested at September 30, 20213,831,850$61.24 
Outstanding at September 30, 2022Outstanding at September 30, 20224,069,431$62.96 
GrantedGranted1,691,36756.73 Granted1,144,59434.27 
VestedVested(962,188)49.10 Vested(798,271)53.72 
ForfeitedForfeited(95,625)69.32 Forfeited(93,688)54.58 
Unvested at June 30, 20224,465,404$61.97 
Outstanding at June 30, 2023Outstanding at June 30, 20234,322,066$57.45 
During the three months ended June 30, 2022 and 2021, the Company recorded $33.7 million and $15.4 million of expense related to RSUs, respectively. During the nine months ended June 30, 2022 and 2021, the Company recorded $83.4 million and $32.5 million of expense related to RSUs, respectively. Such expense is included in stock-based compensation expense in the Company’s Consolidated Statements of Operations and Comprehensive Income (Loss). For RSUs, the expense creates a timing difference, resulting in a deferred tax asset, which is fully reserved by a valuation allowance.
For RSUs, the grant dateThe fair value of the award isRSUs was determined based on the closing price of the Company’s closingcommon stock price aton the grant date, with consideration given to the probability of achieving service and/or performance conditions for performance-based awards. The grant date fair value of the RSUs granted by the Company for
For the three months ended June 30, 2023 and 2022, and 2021 was $42.5the Company recorded $17.8 million and $3.1$33.7 million of expense
20


related to RSUs, respectively. The grant date fair value of the RSUs granted by the Company forFor the nine months ended June 30, 2023 and 2022, and 2021 was $95.2the Company recorded $53.2 million and $112.1$83.4 million of expense related to RSUs, respectively.
As of June 30, 2022, the pre-tax2023, there was $131.6 million of total unrecognized compensation expense for all unvestedcost related to RSUs in the amount of $190.3 million willthat is expected to be recognized in the Company’s results of operations over a weighted averageweighted-average period of 2.72.3 years.

21


NOTE 10. FAIR VALUE MEASUREMENTS
The Company measures its financial assets and liabilities atemploys a fair value hierarchy that prioritizes the inputs to valuation techniques used to measure fair value. FairThe fair value of a financial instrument is defined as the priceamount that would be received to sell an asset or paid to transfer a liability (i.e., exit price) in an orderly transaction between market participants at the measurement date. Additionally,date using the Company is requiredexit price. Accordingly, when market observable data are not readily available, the Company’s own assumptions are used to provide disclosure and categorize assetsreflect those that market participants would be presumed to use in pricing the asset or liability at the measurement date.
Assets and liabilities measuredrecorded at fair value into one of three different levels depending on the assumptions (i.e., inputs) used in the valuation. Level 1 provides the most reliable measure of fair value while Level 3 generally requires significant management judgment. Financial assets and liabilitiesconsolidated balance sheets are classified in their entiretycategorized based on the lowest level of input significantjudgment associated with inputs used to measure their fair values and the fair value measurement. The fair value hierarchy is definedlevel of market price observability, as follows:

Level 1—Valuations are based on unadjusted1    Unadjusted quoted prices are available in active markets for identical assets or liabilities.liabilities as of the reporting date.
Level 2—Valuations2    Pricing inputs are other than quoted prices in active markets, which are based on quotedthe following:
• Quoted prices for similar assets or liabilities in active markets,markets;
• Quoted prices for identical or quoted pricessimilar assets or liabilities in markets that are not active for which significant inputs are observable, eithernon-active markets; or
• Either directly or indirectly.indirectly observable inputs as of the reporting date.
Level 3—Valuations3    Pricing inputs are based on prices or valuation techniques that require inputs that are both unobservable and significant to the overall fair value measurement. Inputs reflect management’s best estimatemeasurement, and the determination of what market participants would usefair value requires significant management judgment or estimation.
In certain cases, inputs used to measure fair value may fall into different levels of the fair value hierarchy. In such cases, the level in valuingthe fair value hierarchy within which the fair value measurement in its entirety falls has been determined based on the lowest level input that is significant to the fair value measurement in its entirety. Thus, a Level 3 fair value measurement may include inputs that are observable (Level 1 or Level 2) and unobservable (Level 3). The Company’s assessment of the significance of a particular input to the fair value measurement in its entirety requires judgment and consideration of factors specific to the asset or liability atliability.
The Company uses prices and inputs that are current as of the measurement date.
17


date, including during periods of market disruption. In periods of market disruption, the ability to observe prices and inputs may be reduced for many instruments. This condition could cause an instrument to be reclassified from Level 1 to Level 2, or from Level 2 to Level 3. The Company recognizes transfers between levels at either the actual date of the event or a change in circumstances that caused the transfer. At June 30, 2023 and September 30, 2022, the Company did not have any financial assets or financial liabilities based on Level 3 measurements.
The following table summarizes fair value measurements at June 30, 2022 and September 30, 2021 forpresents information about the Company’s assets and liabilities measured at fair value on a recurring basis:basis, and indicates the fair value hierarchy of the valuation techniques utilized by the Company:
June 30, 2023
Level 1Level 2Level 3Total
(in thousands)
Financial assets:
U.S. government bonds$27,092 $— $— $27,092 
Municipal securities— 7,033 — 7,033 
Commercial notes— 62,755 — 62,755 
Corporate debt securities— 287,749 — 287,749 
Certificate of deposits— — — — 
Money market instruments49,199 — — 49,199 
22


September 30, 2022
Level 1Level 2Level 3Total
(in thousands)
U.S. government bonds$1,973 $— $— $1,973 
Commercial notes— 41,727 — 41,727 
Corporate debt securities— 271,333 — 271,333 
Certificate of deposits50,000 — — 50,000 
Money market instruments39,262 — — 39,262 
NOTE 11. LIABILITY RELATED TO THE SALE OF FUTURE ROYALTIES
On November 9, 2022, the Company and Royalty Pharma entered into the Royalty Pharma Agreement, pursuant to which Royalty Pharma agreed to pay up to $410.0 million in cash to the Company in consideration for the Company’s future royalty interest in Olpasiran, a small interfering RNA (siRNA) originally developed by the Company and licensed to Amgen in 2016 under the Olpasiran Agreement.
Pursuant to the Royalty Pharma Agreement, Royalty Pharma paid $250.0 million upfront and agreed to pay up to an additional $160.0 million in aggregate one-time milestone payments due if and when the following milestone events occur: (i) $50.0 million on completion of enrollment in the OCEAN Phase 3 clinical trial for Olpasiran, (ii) $50.0 million upon receipt of FDA approval of Olpasiran for an approved indication (reduction in the risk of myocardial infarction, urgent coronary revascularization, or coronary heart disease death in adults with established cardiovascular disease and elevated Lp(a)), and (iii) $60.0 million upon Royalty Pharma’s receipt of at least $70.0 million of royalty payments under the Royalty Pharma Agreement in any single calendar year.
In consideration for the payment of the foregoing amounts under the Royalty Pharma Agreement, Royalty Pharma is entitled to receive all royalties otherwise payable by Amgen to the Company under the Olpasiran Agreement. The Company remains eligible to receive any milestone payments potentially payable by Amgen under the Olpasiran Agreement.
The Company has evaluated the terms of the Royalty Pharma Agreement and concluded in accordance with the relevant accounting guidance that the Company accounted for the transaction as debt and the funding of $250.0 million from Royalty Pharma was recorded as a liability related to the sale of future royalties on its consolidated balance sheets. The Company is not obligated to repay this upfront funding received under the Royalty Pharma Agreement. This liability is amortized over the expected repayment term using an effective interest rate method. The effective interest rate is calculated based on the rate that would enable the debt to be repaid in full over the anticipated life of the arrangement. The interest rate may vary during the term of the agreement depending on a number of factors, including the amount and timing of forecasted net revenues which affects the repayment timing and ultimate amount of repayment. The Company will evaluate the effective interest rate periodically based on its current revenue forecasts utilizing the prospective method. For the three and nine months ended June 30, 2023, the Company recognized non-cash interest expense of $5.2 million and $13.1 million, respectively, on the consolidated statements of operations and comprehensive loss.
NOTE 12. EARNINGS PER SHARE
The following table presents the computation of basic and diluted earnings per share for the nine months ended
23


June 30, 2022:2023 and 2022.
Level 1Level 2Level 3Total
(in thousands)
Classified as Cash and cash equivalents    
Commercial Notes$— $25,336 $— $25,336 
Classified as Marketable Securities    
Marketable securities$— $— $— $— 
Classified as Held to Maturity    
Short-term investments$— $225,490 $— $225,490 
Long-term investments$— $159,540 $— $159,540 
Certificate of deposits$50,000 $— $— $50,000 
Classified as Contingent Consideration    
Contingent consideration$— $— $— $— 
Three Months Ended June 30,Nine Months Ended June 30,
2023202220232022
(in thousands, except per share amounts)
Numerator:
Net loss$(102,946)$(72,046)$(95,596)$(90,552)
Denominator:
Weighted-average basic shares outstanding107,004 105,753 106,597 105,273 
Effect of dilutive securities— — — — 
Weighted-average diluted shares outstanding107,004 105,753 106,597 105,273 
Basic earnings per share$(0.96)$(0.68)$(0.90)$(0.86)
Diluted earnings per share$(0.96)$(0.68)$(0.90)$(0.86)
SeptemberPotentially dilutive securities representing approximately 3,467,000 and 4,024,000 shares of common stock were excluded from the computation of diluted earnings per share for the three and nine months ended June 30, 2021:
Level 1Level 2Level 3Total
(in thousands)
Classified as Marketable Securities    
Marketable securities$126,728 $— $— $126,728 
Classified as Held to Maturity    
Short-term investments$— $57,430 $— $57,430 
Long-term investments$— $196,643 $— $196,643 
Certificate of deposit$50,000 $— $— $50,000 
Classified as Contingent Consideration    
Contingent consideration$— $— $— $— 
2023, respectively, because their effect would have been anti-dilutive.
NOTE 11.
SUBSEQUENT EVENTS
None.
1824


ITEM 2.    MANAGEMENT’S DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND RESULTS OF OPERATIONS
FORWARD-LOOKING STATEMENTS
This Quarterly Report on Form 10-Q contains certain forward-looking statements within the meaning of Section 27A of the Securities Act of 1933, as amended, and Section 21E of the Securities Exchange Act of 1934, as amended, and we intend that such forward-looking statements be subject to the safe harbors created thereby. For this purpose, any statements contained in this Quarterly Report on Form 10-Q except for historical information may be deemed to be forward-looking statements. Without limiting the generality of the foregoing, words such as “may,” “will,” “expect,” “believe,” “anticipate,” “intend,” “plan,” “project,” “could,” “estimate,” “target,” “forecast,”“forecast” or “continue” or the negative of these words or other variations thereof or comparable terminology are intended to identify forward-looking statements. In addition, any statements that refer to projections of ourthe Company’s future financial performance, trends in ourits business, or other characterizations of future events or circumstances are forward-looking statements. These forward-looking statements include, but are not limited to, statements about the initiation, timing, progress and results of the Company’s preclinical studies and clinical trials, and its research and development programs; its expectations regarding the potential benefits of the partnership, licensing and/or collaboration arrangements and other strategic arrangements and transactions the Company has entered into or may enter into in the future; its beliefs and expectations regarding the amount and timing of future milestone, royalty or other payments that could be due to or from third parties under existing agreements; and its estimates regarding future revenues, research and development expenses, capital requirements and payments to third parties.
The forward-looking statements included herein are based on current expectations of ourthe Companys management based on available information and involve a number of risks and uncertainties, all of which are difficult or impossible to predict accurately, and many of which are beyond ourthe Company’s control. In addition, many of these risks and uncertainties may be exacerbated by the COVID-19 pandemic and any worsening of the global business and economic environment as a result. As such, ourthe Company’s actual results and timing of certain events may differ materially from those expressedthe results discussed, projected, anticipated or indicated in any forward-looking statements. Readers should carefully reviewForward-looking statements are not guarantees of future performance and the factors identifiedCompany’s actual results of operations, financial condition and cash flows may differ materially. Factors that may cause or contribute to such differences include, but are not limited to, those discussed in ourmore detail in “Item 1. Business” and Item 1A. Risk Factors” of Part I and “Item 7. Management’s Discussion and Analysis of Financial Condition and Results of Operations” of Part II of the Company’s most recent Annual Report on Form 10-K under the caption “Risk Factors”10-K. Readers should carefully review these risks, as well as the additional risks and uncertainties described in other documents we filethe Company files from time to time with the Securities and Exchange Commission (“SEC”(the “SEC”), including this Quarterly Report on Form 10-Q for the quarter ended June 30, 2022.. In light of the significant risks and uncertainties inherent in the forward-looking information included herein, the inclusion of such information should not be regarded as a representation by usthe Company or any other person that such results will be achieved, and readers are cautioned not to place undue reliance on such forward-looking information. Statements made herein are as of the date of the filing of this Quarterly Report on Form 10-Q with the SEC and should not be relied upon as of any subsequent date. Except as may be required by law, we disclaimthe Company disclaims any intent to revise the forward-looking statements contained herein to reflect events or circumstances after the date hereof or to reflect the occurrence of unanticipated events.
Description of BusinessOVERVIEW
Unless otherwise noted, (1) the term “Arrowhead” refers to Arrowhead Pharmaceuticals, Inc., a Delaware corporation and its Subsidiaries, (2) the terms “Company,” “we,” “us,” and “our,” refer to the ongoing business operations of Arrowhead and its Subsidiaries, whether conducted through Arrowhead or a subsidiary of Arrowhead, (3) the term “Subsidiaries” refers to Arrowhead Madison Inc. (“Arrowhead Madison”), Visirna Therapeutics Inc. ("Visirna"), and Arrowhead Australia Pty Ltd (“Arrowhead Australia”), (4) the term “Common Stock” refers to Arrowhead’s Common Stock, par value $0.001 per share, (5) the term “Preferred Stock” refers to Arrowhead’s Preferred Stock, par value $0.001 per share, and (6) the term “Stockholder(s)” refers to the holders of Arrowhead’s Common Stock.
Overview
Arrowhead Pharmaceuticals, Inc.The Company develops medicines that treat intractable diseases by silencing the genes that cause them. Using a broad portfolio of RNA chemistries and efficient modes of delivery, Arrowheadthe Company’s therapies trigger the RNA interferenceRNAi mechanism to induce rapid, deep and durable knockdown of target genes. RNA interference (“RNAi”)RNAi is a mechanism present in living cells that inhibits the expression of a specific gene, thereby affecting the production of a specific protein. Arrowhead’s RNAi-based therapeutics may leverage this natural pathway of gene silencing. silencing to target and shut down specific disease-causing genes.
The Company has focused its resources on therapeutics that exclusively utilize its high levels of pharmacologic activity in multiple animal models spanning several therapeutic areas. The Company believes that TRiMTM enabled therapeutics offer several potential advantages over prior generation and competing technologies, including: simplified manufacturing and reduced costs; multiple routes of administration including subcutaneous injection and inhaled administration; the ability to target multiple tissue types including liver, lung, muscle, CNS and others; and the potential for improved safety and reduced risk of intracellular buildup, because there are fewer metabolites from smaller, simpler molecules.
The Company’s pipeline includesincludes:
Hypertriglyceridemia - ARO-APOC3 for hypertriglyceridemia,
Dyslipidemia - ARO-ANG3 for dyslipidemia, ARO-ENaC2 for cystic fibrosis, ARO-DUX4 for facioscapulohumeral muscular dystrophy, ARO-COV for the coronavirus that causes COVID-19 and other possible future pulmonary-borne pathogens, ARO-C3 for complement mediated diseases, ARO-RAGE and ARO-MUC5AC for various muco-obstructive or inflammatory pulmonary conditions and ARO-MMP7 for idiopathic pulmonary fibrosis. ARO-HSD for liver
Cardiovascular disease was out-licensed to Glaxosmithkline Intellectual Property (No. 3) Limited (“GSK”) in November 2021. ARO-XDH is being developed for uncontrolled gout under a collaboration agreement with Horizon Therapeutics Ireland DAC (“Horizon”). JNJ-75220795 (ARO-JNJ1) is being developed by Janssen as a potential treatment for patients with non-alcoholic steatohepatitis (NASH).ARO-AAT for liver disease associated with alpha-1 antitrypsin deficiency (“AATD”) was out-licensed to Takeda Pharmaceuticals U.S.A., Inc. (“Takeda”) in October 2020. JNJ-3989 (formerly referred to as ARO-HBV) for chronic hepatitis B virus was out-licensed to Janssen in October 2018.- Olpasiran (formerly referred to as AMG 890 or ARO-LPA) for cardiovascular disease wasARO-LPA, out-licensed to Amgen Inc. (“Amgen”) in 2016. While the Company believes that initial ARO-HIF2 Phase 1 clinical data provides proof of concept for the ability to deliver siRNA to RCC tumors, the Company has decided not to pursue further clinical development of ARO-HIF2 based on a number of factors including the evolving competitive landscape for HIF2 inhibitors.Amgen)
1925


ArrowheadCystic fibrosis - ARO-ENAC2
Muco-obstructive or inflammatory pulmonary conditions - ARO-MUC5AC and ARO-RAGE
Idiopathic pulmonary fibrosis - ARO-MMP7
Non-alcoholic steatohepatitis (NASH) - GSK-4532990 (formerly ARO-HSD, out-licensed to GSK)
Alpha-1 antitrypsin deficiency (AATD) - Fazirsiran (formerly ARO-AAT, a collaboration with Takeda)
Chronic hepatitis B virus - JNJ-3989 (formerly ARO-HBV, out-licensed to Janssen)
Uncontrolled gout - HZN-457 (formerly ARO-XDH, out-licensed to Horizon)
Complement mediated diseases - ARO-C3
Non-alcoholic steatohepatitis (NASH) - ARO-PNPLA3 (formerly JNJ-75220795 or ARO-JNJ1)
Facioscapulohumeral muscular dystrophy - ARO-DUX4
Amyotrophic lateral sclerosis “ALS” (CNS) - ARO-SOD1
The Company operates lab facilities in Madison, Wisconsin and San Diego, California and Madison, Wisconsin, where the Company’sits research and development activities, including the development of RNAi therapeutics, take place. The Company’s principal executive offices are located in Pasadena, California.
DuringThe Company continues to develop other clinical candidates for future clinical trials. Clinical candidates are tested internally and through GLP toxicology studies at outside laboratories. Drug materials for such studies and clinical trials are either manufactured internally or contracted to third-party manufacturers. The Company engages third-party contract research organizations (CROs) to manage clinical trials and works cooperatively with such organizations on all aspects of clinical trial management, including plan design, patient recruiting, and follow up. These outside costs, relating to the preparation for and administration of clinical trials, are referred to as “candidate costs.” As clinical candidates progress through clinical development, candidate costs will increase.
The First Three Quarters of Fiscal 2023 Business Highlights
Key recent developments during the first three quarters of fiscal 2022,2023 included the following:
hosted a Research & Development (R&D) Day on June 1, 2023 to discuss progress of the Company's pipeline of RNAi Therapeutics, at which the following updates were discussed:
ARO-RAGE showed continued dose response with single inhaled dose of 184 mg achieving mean knockdown of 90% and max of 95%;
adipose delivery platform achieved single dose target gene silencing of greater than 90% with six months of duration in non-human primates;
improved hepatic dimer platform achieved equivalent or better knockdown of two target genes with longer duration than monomer mixture in non-human primates;
TRiM™ platform now has potential to address multiple cell types including liver, solid tumors, lung, central nervous system, skeletal muscle, and adipose;
announced progress towards the Company's "20 in 25" goal to grow its pipeline of RNAi therapeutics that leverage the proprietary Targeted RNAi Molecule (TRiM™) platform to a total of 20 clinical stage or marketed products in the year 2025;
presented updated data from the Phase 2 SEQUOIA study of investigational RNAi therapy Fazirsiran in patients with alpha-1 antitrypsin deficiency liver disease which included:
Fazirsiran reduced serum Z-AAT concentration in a dose-dependent manner;
Fazirsiran significantly reduced liver Z-AAT;
Fazirsiran consistently reduced hepatic globule burden;
Fazirsiran treatment reduced histological signs of hepatic inflammation;
50% of the pooled Fazirsiran treated patients showed at least a one-point improvement in METAVIR liver fibrosis versus 38% in the placebo group;
Fazirsiran has been well tolerated to date;
pulmonary function test results (FEV1 and DLCO) for both Fazirsiran and placebo were stable over time with no apparent dose-dependent effects;
updated Phase 2 clinical data were presented at the European Association for the Study of the Liver
26


(EASL) Congress 2023 in an oral presentation titled, “Fazirsiran reduces liver Z-alpha-1 antitrypsin synthesis, decreases globule burden and improves histological measures of liver disease in adults with alpha-1 antitrypsin deficiency: a randomized placebo-controlled phase 2 study”;
presented interim data from the ongoing Phase 2 GATEWAY clinical study of ARO-ANG3 which included:
mean reduction in LDL-C of 48.1% (200mg) and 44.0% (300mg);
ANPTL3 inhibition with ARO-ANG3 also reduced HDL-C, non-HDL-C, and triglycerides, consistent with published human genetic data;
safety and tolerability;
completed enrollment of the Phase 3 PALISADE clinical trial evaluating ARO-APOC3 for treatment of familial chylomicronemia syndrome;
secured stockholder approval to increase authorized common shares to 290,000,000 from 145,000,000 to provide the Company continuedwith additional flexibility to developissue common stock for a variety of general corporate purposes;
announced interim results from ARO-RAGE administration in Part 1 of the ongoing Phase 1/2 study in normal healthy volunteers which included:
reductions in soluble RAGE (sRAGE) as measured in serum after two doses on Day 1 and advance its pipelineDay 29;
duration of pharmacologic effect persisted for at least 6 weeks after the second administration of the 92 mg does with further follow up ongoing;
reduction in sRAGE as measured in bronchoalveolar lavage fluid (BALF) at Day 31 after a single dose;
reduction in in serum sRAGE was observed after a single dose;
the pooled placebo groups experienced a mean sRAGE increase of 8% in BALF and partnered candidatesa mean decrease of 1% serum;
safety and expanded its facilities to support the Company’s growing pipeline. Several key recent developments include:tolerability;
i)expanded TRiMTM platform to include an optimized intrathecal administration for CNS delivery with distribution throughout the brain and in all relevant brain cell types. The first development candidate to utilize this new delivery platform is ARO-SOD1. In June 2023, the Company filed a clinical trial application (CTA) for approval to initiate a Phase 1 clinical study. In preclinical studies, ARO-SOD1 achieved 95% spinal cord tissue mRNA knockdown after a single intrathecal dose in human SOD1 transgenic rats and maintained greater than 80% spinal cord tissue mRNA knockdown three months after a single intrathecal dose in non-human primates;
dosed the first patient in Takeda’s Phase 3 REDWOOD clinical study of Fazirsiran for the treatment of alpha-1 antitrypsin deficiency associated liver diseases, triggering a $40.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023;
dosed the first patient in GSK’s Phase 2b trial of GSK4532990, formerly called ARO-HSD, an investigational RNAi therapeutic for the treatment of patients with non-alcoholic steatohepatitis (NASH), triggering a $30.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023;
announced that the U.S. Food and Drug Administration (FDA) has granted Fast Track designation to ARO-APOC3 for reducing triglycerides in adult patients with familial chylomicronemia syndrome (FCS). ARO-APOC3 was previously granted Orphan Drug designation by the FDA and the European Union;
announced interim results from Part 1 of AROC3-1001, an ongoing Phase 1/2 clinical study of ARO-C3, which included:
a dose-dependent reduction in serum C3, with 88% mean reduction at highest dose tested;
a dose-dependent reduction in AH50, a marker of alternative complement pathway hemolytic activity, with 91% mean reduction at highest dose tested;
duration of pharmacologic effect supportive of quarterly or less frequent subcutaneous dose administration;
safety and tolerability;
received notice from Janssen of its PALISADE study,decision to voluntarily terminate the Janssen Collaboration Agreement
27


between the Company and Janssen. The Company regained full rights to ARO-PNPLA3, formerly called JNJ-75220795, upon termination of the Janssen Collaboration Agreement which took effect on April 7, 2023. ARO-PNPLA3 is in Phase 1 clinical trials that are now being developed by the Company;
initiated dosing in ARO-MMP7-1001 (NCT05537025), a phase 3Phase 1/2a single ascending dose and multiple ascending dose clinical study to evaluate the safety, tolerability, pharmacokinetics, and efficacypharmacodynamics of ARO-APOC3 in adults with familial chylomicronemia syndrome (FCS);
ii)entered intoARO-MMP7, an exclusive license agreement with GSK for ARO-HSD;
iii)Janssen presented clinical data from REEF-1, a Phase 2b study of different combination regimens, including JNJ-73763989 (JNJ-3989), formerly called ARO-HBV, and/or JNJ-56136379 (JNJ-6379), and a nucleos(t)ide analog (NA) for the treatment of chronic hepatitis B virus infection (CHB);
iv)filed for regulatory clearance to begin a Phase 1/2a study of ARO-C3 and subsequently dosed the first subjects in AROC3-1001, a Phase 1/2 clinical study of ARO-C3, the Company’s investigational RNA interference (RNAi)investigational RNAi therapeutic designed to reduce production of complement component 3 (C3) as a potential therapy for various complement mediated diseases;
v)presented additional interim clinical data from AROHSD1001, AROAAT2002, and AROAPOC31001;
vi)completed the purchase of 13 acres of land in the Verona Technology Park in Verona, Wisconsin and held a groundbreaking ceremony on the site. The site is being developed into an approximately 160,000 square foot drug manufacturing facility and an approximately 140,000 square foot laboratory and office facility which will support the Company's process development and analytical activities. The Company also announced that it received awards for up to $16 million in tax increment financing from the city of Verona, and up to $2.5 million in refundable Wisconsin state income tax credits from the Wisconsin Economic Development Corporation (WEDC) as incentives to invest in the local community and create new jobs. Additionally, The Company entered into a lease agreement for a new 144,000 square foot laboratory and office facility in San Diego, California to support discovery activities;
vii)completed enrollment in Phase 2b ARCHES-2 study of investigational ARO-ANG3 for patients with mixed dyslipidemia;
viii)filed for regulatory clearance to initiate Phase 1/2a study of ARO-RAGE for treatment of Asthma;
ix)filed for regulatory clearance to initiate Phase 1/2a study of ARO-MUC5AC for treatment of muco-obstructive lung disease;
x)initiated and dosed the first patients in the Phase 2 GATEWAY clinical study of investigational ARO-ANG3 for the treatment of patients with homozygous familial hypercholersterolemia;
xi)entered into definitive agreements to form a joint venture, Visirna Therapeutics, Inc. (“Visirna”) with Vivo Capital (“Vivo”) through which the Company and Vivo intend to expand the reach of innovative medicines in Greater China;
xii)hosted a pulmonary research & development (R&D) Day to discuss the Company’s emerging pipeline of pulmonary targeted RNA interference (RNAi) therapeutic candidates that leverage its proprietary Targeted RNAi Molecule (TRIMTM) platform, including an announcement of its previously undisclosed candidate designed to reduce expression of matrix metalloproteinase 7 (MMP7) as a potential treatment for idiopathic pulmonary fibrosis (IPF);, in up to 56 healthy volunteers and in up to 21 patients with IPF;
xiii)enrolled the first subject in conjunction with Takeda, a Phase 1 randomized, placebo-controlled trial to assess the safety tolerability, pharmacokinetics and pharmacodynamics of a development-stage medicine, HZN-457 (previously known as ARO-XDH), which is out-licensed to Horizon, triggering a $15.0 million milestone payment to the Company which was paid in the second quarter of fiscal 2023;
enrolled the first subject in Amgen’s Phase 3 trial of Olpasiran, which triggered a $25.0 million milestone payment to the Company, which was paid in the second quarter of fiscal 2023;
entered into the Royalty Pharma Agreement on November 9, 2022, pursuant to which Royalty Pharma paid $250.0 million upfront (See Note 11 — Liability Related to the Sale of Future Royalties of Notes to Consolidated Financial Statements of Part I, “Item 1. Financial Statements.”);
announced top line results from athe SEQUOIA Phase 2 clinicalStudy of Fazirsiran in patients with Alpha-1 Antitrypsin Deficiency-Associated Liver Disease in which;
fibrosis regression was observed in 50% of patients receiving Fazirsiran;
median reductions of 94% of Z-AAT accumulation in the liver and mean reductions of 68% in histologic globule burden were observed;
treatment emergent adverse events were generally well balanced between Fazirsiran and placebo groups;
results were consistent with AROAAT-2002 open-label study (AROAAT-2002) of investigational fazirsiran (TAK-999/ARO-AAT) for the treatment of liver disease associated with alpha-1 antitrypsin deficiency (AATD), and was recentlypreviously published in theThe New England Journal of Medicine (NEJM) and presented in an oral presentation at The International Liver Congress™ 2022 -Medicine.
20


Net loss was $102.9 million for the three months ended June 30, 2023 as compared to $72.0 million for the three months ended June 30, 2022. Net loss was $95.6 million for the nine months ended June 30, 2023 as compared to $90.6 million for the nine months ended June 30, 2022. Net loss per share – diluted was $0.96 for the three months ended June 30, 2023 as compared to $0.68 for the three months ended June 30, 2022. Net loss per share – diluted was $0.90 for the nine months ended June 30, 2023 as compared to $0.86 for the nine months ended June 30, 2022.
The Annual Meeting of the European Association for the Study of the Liver (EASL).
The Company is actively monitoring the ongoing COVID-19 pandemic. The financial resultschanges in net loss for the three and nine months ended June 30, 2022 were not significantly impacted by COVID-19. Operationally, the Company has experienced delays in its earlier stage programs due to a shortage in non-human primates, which are critical to the Company’s preclinical programs. Additionally, the Company has experienced delays in enrollment in its clinical trials. The Company’s operations at its research and development facilities in Madison, Wisconsin and San Diego, California, and its corporate headquarters in Pasadena, California have continued with limited impact, other than for enhanced safety measures and intermittent lab supply shortages. However, the Company cannot predict the impact the progression of COVID-19 will have on future financial and operational results due to a variety of factors, including the ability of the Company’s clinical sites to continue to enroll subjects, the ability of the Company’s suppliers to continue to operate, the continued good health and safety of the Company’s employees and the length and severity of the COVID-19 pandemic.
Net losses were $72.0 million for the three months ended June 30, 2022 as compared to net losses of $29.9 million for the three months ended June 30, 2021. Net losses were $90.6 million for the nine months ended June 30, 2022 as compared to net losses of $77.5 million for the nine months ended June 30, 2021. Net losses per share-diluted were $0.68 for the three months ended June 30, 2022 as compared to net losses per share-diluted of $0.29 for the three months ended June 30, 2022. Net losses per share-diluted were $0.86 for the nine months ended June 30, 2022 as compared to net losses per share-diluted of $0.75 for the nine months ended June 30, 2021. The increase in net losses for the three months ended June 30, 2022 was due to2023 reflect an increase in research and development and general and administrative expenses, which have continued to increase as the Company’s pipeline of candidates has expanded and progressed through clinical trial phases, partially offset by an increase in revenue from the Company’s license and collaboration agreements, primarily from the Takeda License Agreement (as defined below) and the Horizon License Agreement (as defined below).phases.
The Company has strengthened its liquidity and financial position through upfront and milestone payments received under its collaboration agreements, as well as equity financings. Under the terms of the Company’s agreements with Janssen, taken together, the Company has received $175.0 million as an upfront payment, $75.0 million in the form of an equity investment by JJDC in Arrowhead Common Stock, and four milestone payments totaling $70.0 million. Under the terms of the Company’s agreements with Amgen, the Company has received $35.0 million in upfront payments, $21.5 million in the form of an equity investment by Amgen in the Company’s Common Stock and $30.0 million in milestone payments. The Takeda License Agreement resulted in a $300.0 million upfront payment, and the Horizon License Agreement resulted in a $40.0 million upfront payment. Finally, the GSK License Agreement resulted in an upfront payment of $120.0 million, which was received in January 2022.The Company had $139.4$105.3 million of cash, and cash equivalents $277.1and restricted cash, $346.4 million in short-term investments, $165.9$42.8 million of long termlong-term investments and $751.8$795.9 million of total assets as of June 30, 2022,2023, as compared to $184.4$108.0 million of cash, and cash equivalents $126.7 million of marketable securities, $56.6and restricted cash, $268.4 million in short-term investments, $245.6$105.9 million of long termlong-term investments and $710.1$691.9 million of total assets as of September 30, 2021.2022. Based upon the Company’s current cash and investment resources and operating plan, the Company expects to have sufficient liquidity to fund operations for at least the next twelve months.
Critical Accounting Policies and Estimates
There have been no significant changes to the significantCompany’s critical accounting policiesestimates disclosed in the Company’s most recent Annual Report on Form 10-K.10-K for the fiscal year ended September 30, 2022, except the Takeda revenue recognition described in Note 2 — Collaboration and License Agreements of Notes to Consolidated Financial Statements of Part I, “Item 1. Financial Statements.”
28

Results of Operations

RESULTS OF OPERATIONS
The following data summarizes ourthe Company’s results of operations for the following periods indicated:
Three Months Ended June 30,
20222021
(in thousands, except per share
amounts)
Revenues$32,412 $45,891 
Operating income (loss)$(72,909)$(31,868)
Net income (loss)$(72,046)$(29,924)
Net income (loss) per share-diluted(0.68)(0.29)
21


Nine Months Ended June 30,
20222021
(in thousands, except per share
amounts)
Revenues$211,656 $100,004 
Operating income (loss)$(94,677)$(84,153)
Net income (loss)$(90,552)$(77,474)
Net income (loss) per share-diluted(0.86)(0.75)
The increase in revenue for the nine months ended June 30, 2022 compared to the nine months ended June 30, 2021 was driven by the revenue recognized from the GSK, Takeda and Horizon License Agreements.
Three Months Ended June 30,Nine Months Ended June 30,
2023202220232022
(in thousands, except per share amounts)
Revenues$15,825 $32,412 $224,638 $211,656 
Operating loss$(102,703)$(72,909)$(96,672)$(94,677)
Net loss attributable to Arrowhead Pharmaceuticals, Inc.$(102,946)$(72,046)$(95,596)$(90,552)
Net loss per share-diluted$(0.96)$(0.68)$(0.90)$(0.86)
Revenue
Total revenue for the three months ended June 30, 2022 and 2021 was $32.42023 decreased to $15.8 million, and $45.9 million, respectively.or 51.2% from the same period of 2022. Total revenue for the nine months ended June 30, 20222023 increased to $224.6 million, or 6.1% from the same period of 2022. The changes were primarily driven by the revenue recognition associated with GSK, Horizon, Takeda and 2021 was $211.7 millionAmgen license agreements, as discussed below. The Company has evaluated each agreement in accordance with FASB Topic 808–Collaborative Arrangements and $100.0 million, respectively. Topic 606-Revenue for Contracts from Customers. See Note 2 — Collaboration and License Agreements to Consolidated Financial Statements of Part I, “Item 1. Financial Statements” for more information on revenue recognized under the nine months ended June 30, 2022 is primarily related tocollaboration and license agreements.
GSK
At the recognitioninception of the $120.0 million of revenue associated with the upfront payment received from GSK under the GSK License Agreement, in the second quarter of 2022.
Amgen Inc.
On September 28, 2016, the Company entered into two collaboration and license agreements and a common stock purchase agreement with Amgen. Under the Second Collaboration and License Agreement (the “Olpasiran Agreement”), Amgen has received a worldwide, exclusive license to Arrowhead’s novel RNAi Olpasiran (previously referred to as AMG 890 or ARO-LPA) program. These RNAi molecules are designed to reduce elevated lipoprotein(a), which is a genetically validated, independent risk factor for atherosclerotic cardiovascular disease. Under the prior collaboration and license agreement (the “First Collaboration and License Agreement” or the “ARO-AMG1 Agreement”), Amgen received an option to a worldwide, exclusive license for ARO-AMG1, an RNAi therapy for an undisclosed genetically validated cardiovascular target. Under both agreements, Amgen is wholly responsible for clinical development and commercialization. Under the terms of the agreements taken together, the Company has received $35.0 million in upfront payments, $21.5 million in the form of an equity investment by Amgen in the Company’s Common Stock, and $30.0 million in milestone payments, and may receive up to an additional $400.0 million in remaining development, regulatory and sales milestone payments. The Company is further eligible to receive up to low double-digit royalties for sales of products under the Olpasiran Agreement. In July 2019, Amgen informed the Company that it would not be exercising its option for an exclusive license for ARO-AMG1, and as such, there will be no further milestone or royalty payments under the ARO-AMG1 Agreement.
The Company has evaluated these agreements in accordance with FASB Topics 808 – Collaboration Arrangements and 606 - Revenue for Contracts from Customers. The Company has substantially completed its performance obligations under the Olpasiran Agreement and the ARO-AMG1 Agreement. Future milestones and royalties achieved will be recognized in their entirety when earned. In July 2020, Amgen initiated a Phase 2 clinical study of Olpasiran, which resulted in a $20.0 million milestone payment to the Company. During the three and nine months ended June 30, 2022 and 2021, the Company recognized $0 and $0 of revenue associated with its agreement with Amgen, respectively.As of June 30, 2022, there were $0 in contract assets recorded as accounts receivable and $0 contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.
Janssen Pharmaceuticals, Inc.
On October 3, 2018, the Company entered into a License Agreement (the “Janssen License Agreement”) and a Research Collaboration and Option Agreement (the “Janssen Collaboration Agreement”) with Janssen, part of the Janssen Pharmaceutical Companies of Johnson & Johnson. The Company also entered into a stock purchase agreement with JJDC (“JJDC Stock Purchase Agreement”). Under the Janssen License Agreement, Janssen has received a worldwide, exclusive license to the Company’s JNJ-3989 (ARO-HBV) program, the Company’s third-generation subcutaneously administered RNAi therapeutic candidate being developed as a potential therapy for patients with chronic hepatitis B virus infection. Beyond the Company’s Phase 1/2 study of JNJ-3989 (ARO-HBV), which the Company was responsible for completing, Janssen is wholly responsible for clinical development and commercialization of JNJ-3989. Under the Janssen
22


Collaboration Agreement, Janssen was able to select three new targets against which Arrowhead would develop clinical candidates. These candidates were subject to certain restrictions and do not include candidates that already were in the Company’s pipeline. The Company was obligated to perform discovery, optimization and preclinical research and development, entirely funded by Janssen, which on its own or in combination with Janssen development work, would have been sufficient to allow the filing of a U.S. Investigational New Drug Application or equivalent, at which time Janssen would have the option to take an exclusive license. If the option was exercised, Janssen would have been wholly responsible for clinical development and commercialization of each optioned candidate. Under the terms of the agreements taken together, the Company has received $175.0 million as an upfront payment, $75.0 million in the form of an equity investment by JJDC in Arrowhead Common Stock under the JJDC Stock Purchase Agreement, and milestone and option payments totaling $73.0 million, and the Company may receive up to $1.6 billion in development and sales milestones payments for the Janssen License Agreement, and up to $0.6 billion in development and sales milestone payments for the remaining target covered under the Janssen Collaboration Agreement. The Company is further eligible to receive tiered royalties on product sales up to mid-teens under the Janssen License Agreement and up to low teens under the Janssen Collaboration Agreement. During the three months ended June 30, 2022, Janssen’s option period expired unexercised for two of the three candidates (ARO-JNJ2 and ARO-JNJ3) under the Janssen Collaboration Agreement.
The Company has evaluated these agreements in accordance with FASB Topics 808 – Collaboration Arrangements and 606 - Revenue for Contracts from Customers. At the inception of these agreements, the Company identified one distinct performance obligation. Regarding the Janssen License Agreement, theThe Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibility to complete the Phase 1/2 study of JNJ-3989 (ARO-HBV) and the Company’s responsibility to ensure certain manufacturing of JNJ-3989 (ARO-HBV) drug product is completed and delivered to Janssen (the “Janssen“GSK R&D Services”). Due to the specialized and unique nature of these Janssenthe GSK R&D Services and their direct relationship with the license, the Company determined that these deliverables representrepresented one distinct bundle and, thus, one performance obligation. The Company also determined that Janssen’s option to requireBeyond the GSK R&D Services, which are the responsibility of the Company, to develop up to three new targets is not a material rightGSK will be responsible for managing future clinical development and thus, not a performance obligation at the onset of the agreement. The consideration for this option is accounted for separately.commercialization in its territory.
The Company determined the initial transaction price totaled approximately $252.7$120.0 million, which includesincluding the upfront payment, which was collected in January 2022 (see Note 2 — Collaboration and License Agreements to Consolidated Financial Statements of Part I, “Item 1. Financial Statements” for more information on revenue recognized under the premium paid by JJDC for its equity investment in theGSK License Agreement). The Company two $25.0 million milestone payments relatedhas excluded any future estimated milestones or royalties from this transaction price to JNJ-3989 (ARO-HBV), and estimated payments for reimbursable Janssen R&D Services to be performed.date. The Company has allocated the total $252.7$120.0 million initial transaction price to its one distinct performance obligation for the JNJ-3989 (ARO-HBV)GSK-4532990 license and the associated JanssenGSK R&D Services. TheAs the Company has recognized this transaction price in its entirety as of September 30, 2021, ascompleted its performance obligations were substantially completed. Future milestones and royalties achieved will beobligation related to this agreement, the upfront payment of $120.0 million was fully recognized in their entirety when earned. Duringduring the threesix months ended June 30, 2022 and 2021,March 31, 2022. Further, GSK dosed the Company recognized approximately $0 and $0 of revenue associated with this performance obligation, respectively. During the nine months ended June 30, 2022 and 2021, the Company recognized approximately $0 and $20.2 million of revenue associated with this performance obligation, respectively. As of June 30, 2022, there were $0 in contract assets recorded as accounts receivable, and $0 of contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.
The Company has conducted its discovery, optimization and preclinical research and development of JNJ-75220795 (ARO-JNJ1), ARO-JNJ2, and ARO-JNJ3 under the Janssen Collaboration Agreement. All costs and labor hours spent by the Company have been entirely funded by Janssen. During the three months ended June 30, 2022, Janssen’s option period expired unexercised for two of the three candidates (ARO-JNJ2 and ARO-JNJ3) under the Janssen Collaboration Agreement as reported in the second quarter of 2022. During the three months ended June 30, 2022 and 2021, the Company recognized $0.0 million and $0.2 million of revenue associated with these efforts, respectively. During the nine months ended June 30, 2022 and 2021, the Company recognized $0.1 million and $0.5 million of revenue associated with these efforts, respectively. In May 2021, Janssen exercised its option right for ARO-JNJ1, which resultedfirst patient in a $10.0Phase 2b trial in March 2023, triggering a $30.0 million milestone payment to the Company. This $10 million milestone paymentCompany which was recognized entirely duringpaid in the three months endedthird quarter of fiscal 2023.
Horizon
On June 30, 2021.As of June 30, 2022, there were $0 of contract assets recorded as accounts receivable18, 2021, Horizon and $0 of contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.
Takeda Pharmaceuticals U.S.A., Inc.
On October 7, 2020, the Company entered into an Exclusivethe Horizon License and Co-funding agreement (the “Takeda License Agreement”) with Takeda. UnderAgreement. At the Takedainception of the Horizon License Agreement, Takedathe Company identified one distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibilities to conduct all activities through the preclinical stages of development of HZN-457 (the “Horizon R&D Services”). Due to the specialized and unique nature of these Horizon R&D Services and their direct relationship with the license, the Company determined that these deliverables represented one distinct bundle and, thus, one performance obligation. Beyond the Horizon R&D Services, which are the responsibility of the Company, Horizon is responsible for managing future clinical development and commercialization of HZN-457.
The Company determined the initial transaction price totaled $40.0 million, including the upfront payment (see Note 2 — Collaboration and License Agreements to Consolidated Financial Statements of Part I, “Item 1. Financial Statements” for more information on revenue recognized under the Horizon License Agreement). The Company has excluded any future estimated milestones or royalties from this transaction price to date. The Company allocated the total $40.0 million initial transaction price to its one distinct performance obligation for the HZN-457 license and the associated Horizon R&D Services. Revenue was recognized on a straight-line basis over the timeframe for completing the Horizon R&D Services. The Company will co-developdetermined that the Company’s ARO-AAT program,straight-line basis was appropriate as its efforts were expended evenly over the Company’s second-generation subcutaneously administered RNAi therapeuticcourse of completing its performance obligation. Further, Horizon enrolled the first subject in December 2022 in a Phase 1
2329


candidate being developed asrandomized, placebo-controlled trial to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of HZN-457, triggering a treatment for liver disease associated with alpha-1 antitrypsin deficiency. Within the United States, ARO-AAT, if approved, will be co-commercialized under a 50/50 profit sharing structure. Outside the United States, Takeda will lead the global commercialization strategy and will receive an exclusive license$15.0 million milestone payment to commercialize ARO-AAT, while the Company will be eligible to receive tiered royaltieswhich was paid in the second quarter of 20% to 25% on net sales. In January 2021,fiscal 2023.
Takeda
On October 7, 2020, Takeda and the Company received $300.0 million as an upfront payment and is eligible to receive potential development, regulatory and commercial milestones of up to $740.0 million.
The Company has evaluatedentered into the Takeda License Agreement in accordance with FASB Topics 808 – Collaborative Arrangements and 606 - Revenue for Contracts from Customers.Agreement. At the inception of the Takeda License Agreement, the Company identified one distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services including the Company’s responsibilities to complete the initial portion of the SEQUOIA study, to complete the ongoing Phase 2 AROAAT2002 study and to ensure certain manufacturing of ARO-AATFazirsiran drug product is completed and delivered to Takeda (the “Takeda R&D Services”). Due to the specialized and unique nature of these Takeda R&D Services and their direct relationship with the license, the Company determined that these deliverables represent one distinct bundle and, thus, one performance obligation. Beyond the Takeda R&D Services, which are the responsibility of the Company, Takeda will be responsible for managing future clinical development and commercialization outside the United States. Within the United States, the Company will also participate in co-development and co-commercialization efforts and will co-fund these efforts with Takeda as part of the 50/50 profit sharing structure within the United States. The Company considers the collaborative activities, including the co-development and co-commercialization, to be a separate unit of account within Topic 808, and as such, these co-funding amounts will beare recorded as Researchresearch and Development Expensesdevelopment expenses or Generalgeneral and Administrative Expenses,administrative expenses, as appropriate.
The Company determined the initial transaction price totaled $300.0 million, which includes the upfront payment. The Company has excluded any future milestones or royalties from this transaction price to date. The Company has allocated the total $300.0 million initial transaction price to its one distinct performance obligation for the ARO-AATFazirsiran license and the associated Takeda R&D Services. Revenue will beis recognized using a proportional performance method (based on actual patient visits completed versus total estimated visits completed for the ongoing SEQUOIA and AROAAT2002 clinical studies). RevenueSee Note 2 — Collaboration and License Agreements to Consolidated Financial Statements of Part I, “Item 1. Financial Statements” for more information on revenue recognized under the three months ended June 30, 2022Takeda License Agreement. The Company previously expected these clinical trials to extend to September 2025 in order to demonstrate long term safety and 2021 was $25.5 million and $35.7 million, respectively. Revenue forefficacy in the nine months ended June 30, 2022 and 2021 was $67.1 million and $69.3 million, respectively. As of June 30, 2022, there were $0 in contract assets recorded as accounts receivable, $71.0 million in contract liabilities recorded as deferred revenue and $71.2 million in contract liabilities recorded as deferred revenue, netopen label extension (OLE) part of the current portion,studies; however, Takeda now intends to initiate a new OLE study available to patients participating in these Phase 2 studies that initiated in July 2023. Based on this new information, patients enrolled in the SEQUOIA and $7.8 million in contract liabilities recorded as accrued expenses. The $7.8 million in accrued expenses was primarily driven by co-developmentAROAAT2002 studies are expected to complete their Phase 2 study visits between June 2023 and co-commercialization activities.
Horizon Therapeutics Ireland DAC
On June 18, 2021,2024, shortening the Company’s performance obligation. As a result, effective the second quarter of fiscal 2023, the Company entered into the Horizon License Agreement with Horizon. Under the Horizon License Agreement, Horizon received a worldwide exclusive license for ARO-XDH, a previously undisclosed discovery-stage investigational RNAi therapeutic being developed by the Company as a potential treatment for people with uncontrolled gout. The Company will conduct all activities through the preclinical stages of development of ARO-XDH, and Horizon will be wholly responsible for clinical development and commercialization of ARO-XDH. In July 2021, the Company received $40 million as an upfront payment and is eligible to receive up to $660 million in potential development, regulatory and sales milestones. The Company is also eligible to receive royalties in the low- to mid-teens range on net product sales.
The Company has evaluated the Horizon License Agreement in accordance with FASB Topics 808 – Collaborative Arrangements and 606 - Revenue for Contracts from Customers. At the inceptionchanged its estimates of the Horizon License Agreement, the Company identified one distinctrevenue recognition to better reflect these newly estimated proportional performance obligation.periods. The Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibilities to conduct all activities through the preclinical stages of development of ARO-XDH (the “Horizon R&D Services”). Due to the specialized and unique natureeffect of these Horizon R&D Services and their direct relationship with the license, the Company determined that these deliverables represented one distinct bundle and, thus, one performance obligation. Beyond the Horizon R&D Services, which are the responsibility of the Company, Horizon will be responsiblechanges in estimates resulted in accelerated revenue by $61.4 million, or $0.58 per share (diluted) for managing future clinical development and commercialization of ARO-XDH.
The Company determined the initial transaction price totaled $40.0 million, including the upfront payment. The Company has excluded any future estimated milestones or royalties, from this transaction price to date. The Company will allocate the total $40.0 million initial transaction price to its one distinct performance obligation for the ARO-XDH license
24


and the associated Horizon R&D Services. Revenue will be recognized on a straight-line basis over the estimated timeframe for completing the Horizon R&D Services. The Company determined that the straight-line basis was appropriate as its efforts will be expended evenly over the course of completing its performance obligation. Revenue for the three months ended June 30, 2022 and 2021 was $6.7 million and $0, respectively. Revenue for the nine months ended June 30, 2022 and 2021 was $20.0 million and $0, respectively. As of June 30, 2022, there were $0 million in contract assets recorded as accounts receivable, $13.3 million in contract liabilities recorded as deferred revenue.
The Company has manufactured ARO-XDH material for Horizon in furtherance of the research plan entered into pursuant to the Horizon License Agreement, for which the Company has been reimbursed for its costs. During the nine months ended June 30, 2022 and 2021, the Company recognized $1.3 million and $0 with these efforts, respectively. As of June 30, 2022, there were $0.0 million of contract assets recorded as accounts receivable and $0 of contract liabilities recorded as current deferred revenue on the Company’s Consolidated Balance Sheets.
Glaxosmithkline Intellectual Property (No. 3) Limited
On November 22, 2021, the Company entered into an Exclusive License Agreement (the “GSK License Agreement”) with GSK. Under the GSK License Agreement, GSK has received an exclusive license for ARO-HSD, the Company’s investigational RNAi therapeutic being developed as a treatment for patients with alcohol-related and nonalcohol related liver diseases, such as nonalcoholic steatohepatitis (NASH). The exclusive license is worldwide with the exception of greater China, for which the Company retained rights to develop and commercialize. Beyond the Company’s Phase 1/2 study of (ARO-HSD), which the Company is responsible for completing, GSK is wholly responsible for clinical development and commercialization of ARO-HSD in its territory. Under the terms of the agreement, the Company has received an upfront payment of $120 million and is eligible for additional payments of $30 million at the start of Phase 2 and $100 million upon achieving a successful Phase 2 trial readout and the first patient dosed in a Phase 3 trial. Furthermore, should the Phase 3 trial read out positively, and the potential new medicine receives regulatory approval in major markets, the deal provides for commercial milestone payments to the Company of up to $190 million at first commercial sale, and up to $590 million in sales-related milestone payments. The Company is further eligible to receive tiered royalties on net product sales in a range of mid-teens to twenty percent.
The Company has evaluated the GSK License Agreement in accordance with FASB Topics 808 – Collaborative Arrangements and 606 - Revenue for Contracts from Customers. At the inception of the GSK License Agreement, the Company identified one distinct performance obligation. The Company determined that the key deliverables included the license and certain R&D services, including the Company’s responsibility to complete the Phase 1/2 study, (the “GSK R&D Services”). Due to the specialized and unique nature of these GSK R&D Services and their direct relationship with the license, the Company determined that these deliverables represented one distinct bundle and, thus, one performance obligation. Beyond the GSK R&D Services, which are the responsibility of the Company, GSK will be responsible for managing future clinical development and commercialization in its territory.
The Company determined the initial transaction price totaled $120.0 million, including the upfront payment. The $120.0 million upfront payment was collected in January 2022. The Company has excluded any future estimated milestones or royalties from this transaction price to date. The Company has allocated the total $120.0 million initial transaction price to its one distinct performance obligation for the ARO-HSD license and the associated GSK R&D Services. As the Company has completed its performance obligation related to this agreement, the upfront payment of $120.0 million will be fully recognized aseach of the three and nine months ended June 30, 2022. Revenue for the nine months ended June 30, 2022 and 2021 was $120.02023. There were $16.9 million and $0, respectively. As of June 30, 2022, there were $0 in contract assets recorded as accounts receivable, $0 in contract liabilities recorded as deferred revenue.revenue, which was classified as current as of June 30, 2023.

In March 2023, Takeda dosed the first patient in the Phase 3 REDWOOD clinical study of Fazirsiran, triggering a $40.0 million milestone payment to the Company which was paid in the third quarter of fiscal 2023.
Joint VentureAmgen Inc. (“Amgen”)
On September 28, 2016, Amgen and License Agreement with Visirna Therapeutics, Inc.

On April 25, 2022, the Company entered into two collaboration and license agreements and a Licensecommon stock purchase agreement. Under the Olpasiran Agreement, (the “Visirna License Agreement”) with Visirna Therapeutics, Inc. (“Visirna”), pursuant to which VisirnaAmgen received ana worldwide, exclusive license to develop, manufacturethe Company’s novel RNAi Olpasiran program. Olpasiran is designed to reduce elevated lipoprotein(a), which is a genetically validated, independent risk factor for atherosclerotic cardiovascular disease. Amgen is wholly responsible for clinical development and commercialize four of Arrowhead’s RNAi-based investigational cardiobolic medicinescommercialization. The Company has substantially completed its performance obligations under the Olpasiran Agreement.
Further, in Greater China (includingNovember 2022, Royalty Pharma and the People’s Republic of China, Hong Kong, Macau and Taiwan).Pursuant to a Share Purchase AgreementCompany entered into simultaneously with the Visirna License Agreement (the “Visirna SPA”), the Company acquired a majority stake in Visirna (after accounting for shares reserved for Visirna’s employee stock ownership plan) as partialRoyalty Pharma Agreement. In consideration for the Visirna License Agreement.Under the Visirna SPA, entities affiliated with Vivo Capital (“Vivo”) also acquired a minority stake in Visirna in exchange for $60 million in upfront capital to support the operations of Visirna.As further considerationpayments under the Visirna LicenseRoyalty Pharma Agreement, Royalty Pharma is entitled to receive all royalties otherwise payable by Amgen to the Company is alsounder the Olpasiran Agreement. The Company remains eligible to receive potential royalties on commercial sales.any milestone payments potentially payable by Amgen under the Olpasiran Agreement.
In December 2022, Amgen enrolled the first subject in its Phase 3 trial of Olpasiran, which triggered a $25.0 million milestone payment to the Company which was paid in the second quarter of fiscal 2023. The Company is further eligible to receive up to an additional $535.0 million in aggregate development, regulatory, and sales milestone payments from Amgen and Royalty Pharma.

2530



The Company has determined that Visirna is a variable interest entity upon its formation and as of June 30, 2022, and it has been determined that Arrowhead is the primary beneficiary; accordingly, Arrowhead consolidates the financial statements of Visirna.
Operating Expenses
The analysis below details the operating expenses and discusses the expenditures of the Company within the major expense categories. Certain reclassifications have been made to prior-period operating expense categories to conform to the current period presentation. For purposes of comparison, the amounts for the three and nine months ended June 30, 20222023 and 20212022 are shown in the tables below.
Research and Development (R&D) Expenses
R&D expenses are related to the Company’s research and development discovery efforts and related programcandidate costs, which are comprised primarily of outsourced costs related to the manufacturing of clinical supplies, toxicity/efficacy studies and clinical trial expenses. Internal costs primarily relate to discovery operations at ourthe Company’s research facilities in Madison, Wisconsin and San Diego, California and Madison, Wisconsin, including facility costs and laboratory-related expenses. Salaries and stock compensation expense consist of salary, bonuses, payroll taxes and related benefits and stock compensation for our R&D personnel. Depreciation and amortization expense consist of depreciation on lab equipment and leasehold improvements at our research facilities. We doThe Company does not separately track R&D expenses by individual research and development projects, includingor by individual drug candidates. The Company operates in a cross-functional manner across projects and does not separately allocate facilities-related costs, candidate costs, discovery costs, compensation expenses, depreciation and amortization expenses, and other expenses forrelated to research and development activities.
The following table provides details of research and development expenses for the periods indicated:
(table below in thousands)
(in thousands)Three Months Ended
June 30, 2023
% of
Expense
Category
Three Months Ended
June 30, 2022
% of
Expense
Category
Increase (Decrease)
$%
Candidate costs$41,209 44 %$31,732 44 %$9,477 30 %
R&D discovery costs20,253 21 %15,081 21 %5,172 34 %
Salaries16,632 18 %11,243 16 %5,389 48 %
Facilities related4,810 %3,827 %983 26 %
Total research and development expense, excluding non-cash expense$82,904 88 %$61,883 86 %$21,021 34 %
Stock compensation8,982 %8,098 11 %884 11 %
Depreciation and amortization2,871 %2,200 %671 31 %
Total research and development expense$94,757 100 %$72,181 100 %$22,576 31 %
Three Months Ended June 30, 2022
% of
Expense
Category
Three Months Ended June 30, 2021
% of
Expense
Category
Increase (Decrease)
$%
(in thousands)(in thousands)Nine Months Ended
June 30, 2023
% of
Expense
Category
Nine Months Ended
June 30, 2022
% of
Expense
Category
Increase (Decrease)
$%
Candidate costsCandidate costs$110,079 43 %$101,789 47 %$8,290 %
R&D discovery costsR&D discovery costs50,377 20 %40,347 19 %10,030 25 %
SalariesSalaries$11,243 16 %$8,518 14 %$2,725 32 %Salaries47,725 19 %33,641 16 %14,084 42 %
Facilities relatedFacilities related$3,827 %$1,306 %$2,521 193 %Facilities related11,601 %7,644 %3,957 52 %
Candidate costs$31,732 44 %$31,468 54 %$264 %
R&D discovery costs$15,081 21 %$10,082 17 %$4,999 50 %
Total research and development expense, excluding non-cash expenseTotal research and development expense, excluding non-cash expense$61,883 86 %$51,374 87 %$10,509 20 %Total research and development expense, excluding non-cash expense$219,782 87 %$183,421 86 %$36,361 20 %
Stock compensationStock compensation$8,098 11 %$6,530 11 %$1,568 24 %Stock compensation26,129 10 %23,958 11 %2,171 %
Depreciation/amortization$2,200 %$1,421 %$779 55 %
Depreciation and amortizationDepreciation and amortization7,422 %6,551 %871 13 %
Total research and development expenseTotal research and development expense$72,181 100 %$59,325 100 %$12,856 22 %Total research and development expense$253,333 100 %$213,930 100 %$39,403 18 %
26


Nine Months Ended June 30, 2022
% of
Expense
Category
Nine Months Ended June 30, 2021
% of
Expense
Category
Increase (Decrease)
$%
Salaries$33,641 16 %$25,375 18 %$8,266 33 %
Facilities related7,644 %$4,456 %$3,188 72 %
Candidate costs101,789 47 %$67,152 48 %$34,637 52 %
R&D discovery costs40,347 19 %$20,295 14 %$20,052 99 %
Total research and development expense, excluding non-cash expense$183,421 86 %$117,278 83 %$66,143 56 %
Stock compensation$23,958 11 %$18,421 13 %$5,537 30 %
Depreciation/amortization$6,551 %$4,877 %$1,674 34 %
Total research and development expense$213,930 100 %$140,576 100 %$73,354 52 %
Salaries expenseCandidate costs increased by $2,725,000 from $8,518,000 during$9.5 million, or 30%, for the three months ended June 30, 2021 to $11,243,000 during the current period. Salaries expense increased by $8,266,000 from $25,375,000 during2023 and $8.3 million, or 8%, for the nine months ended June 30, 20212023 compared to $33,641,000 during the current period.same period of 2022. This increase iswas primarily due to the additional progression of the Company’s pipeline of candidates into and through clinical trials, which resulted in higher outsourced clinical trial, toxicity study and manufacturing costs.
R&D discovery costs increased $5.2 million, or 34%, for the three months ended June 30, 2023 and $10.0 million, or 25%, for the nine months ended June 30, 2023 compared to the same period of 2022. This increase was due to the growth of the Company’s discovery efforts and continued advancement into novel therapeutic areas and tissue types.
Salaries and stock compensation expense consist of salary, bonuses, payroll taxes, related benefits and stock compensation for the Company’s R&D personnel. The increases in salaries and stock comp expenses for the nine months ended June 30, 2023 were primarily due to an increase in R&D headcount that has occurred as the Company has expanded its pipeline of candidates. We anticipate thiscandidates, in addition to annual salary increases. Stock compensation expense was based upon the valuation of
31


stock options and restricted stock units granted to continueemployees and directors.
Facilities-related expense included lease costs for the Company’s research and development facilities in San Diego, California and Madison, Wisconsin. Facilities-related costs increased $1.0 million, or 26%, for the three months ended June 30, 2023 and $4.0 million, or 52%, for the nine months ended June 30, 2023 compared to the same period of 2022. This increase was mainly due to the additional lease expense as we continue to expand our pipeline of candidates and increase headcount to support ourthe Company expands discovery efforts to identify new drug candidates, in additioncandidates.
Depreciation and amortization expense, a non-cash expense, relates to inflationary pressures indepreciation on lab equipment and leasehold improvements at the labor market.facilities.
Facilities expense increased by $2,521,000 from $1,306,000 during the three months ended June 30, 2021 to $3,827,000 during the current period. Facilities expense increased by $3,188,000 from $4,456,000 during the nine months ended June 30, 2021 to $7,644,000 during the current period. This category includes rental costs for our research and development facilities in Madison, Wisconsin and San Diego, California. We expect this expense to continue to increase as we continue to build out our manufacturing capabilities to support our discovery efforts to identify new drug candidates. The increase in facilities expense is due to the recognition of rent expense from the San Diego research facility.
Candidate costs increased by $264,000 from $31,468,000 during the three months ended June 30, 2021 to $31,732,000 during the current period. Candidate costs increased by $34,637,000 from $67,152,000 during the nine months ended June 30, 2021 to $101,789,000 during the current period. This increase is primarily due to the progression of our pipeline of candidates into and through clinical trials, which results in higher outsourced clinical trial, toxicity study and manufacturing costs. For example, our cardiometabolic candidates, ARO-ANG3 and ARO-APOC3, have advanced into phase 2 and phase 3 clinical trials. We anticipateCompany anticipates these R&D expenses to continue to increase as ourits pipeline of candidates grows and progresses to later phase clinical trials, in addition to unforeseen inflationary pressure on goods and services.
R&D discovery costs increased by $4,999,000 from $10,082,000 duringservices and the three months ended June 30, 2021 to $15,081,000 in the current period. R&D discovery costs increased by $20,052,000 from $20,295,000 during the nine months ended June 30, 2021 to $40,347,000 in the current period. This increase is primarily due to the growth of our discovery efforts, including the addition of our research facility in San Diego. We anticipate this expense to continue to increase as we increase headcount to support our discovery efforts to identify new drug candidates.
Stock compensation expense, a non-cash expense, increased by $1,568,000 from $6,530,000 during the three months ended June 30, 2021 to $8,098,000 during the current period. Stock compensation expense, a non-cash expense, increased by $5,537,000 from $18,421,000 during the nine months ended June 30, 2021 to $23,958,000 during the current period. Stock compensation expense is based upon the valuation of stock options and restricted stock units granted to employees, directors and certain consultants. Many variables affect the amount expensed, including the Company’s stock price on the date of the grant, as well as other assumptions. The increase in the expense in the current period is primarily due to the increased headcount discussed above. We generally expect future stock compensation expense to continue to increase as our headcount continues to increase to support our clinical pipeline.
Depreciation and amortization expense, a non-cash expense, increased by $779,000 from $1,421,000 during the three months ended June 30, 2021 to $2,200,000 during the current period. Depreciation and amortization expense, a non-cash expense, increased by $1,674,000 from $4,877,000 during the nine months ended June 30, 2021 to $6,551,000 during the current period. The majority of depreciation and amortization expense relates to depreciation on lab equipment and
27


leasehold improvements at our Madison and San Diego research facilities. The increase in depreciation and amortization expense is due to an increase in laboratory equipment and leasehold improvements. We expect this amount to increase in the future as we continue to purchase additional lab equipment and increase our footprint expansions to support our growing pipeline.labor market.
General & Administrative Expenses
The following table provides details of ourthe Company’s general and administrative expenses for the periods indicated:
(table below in thousands)
(in thousands)Three Months Ended
June 30, 2023
% of
Expense
Category
Three Months Ended
June 30, 2022
% of
Expense
Category
Increase (Decrease)
$%
Salaries$5,063 21 %$3,175 10 %$1,888 59 %
Professional, outside services, and other5,987 25 %3,568 11 %2,419 68 %
Facilities related1,352 %703 %649 92 %
Total general & administrative expense, excluding non-cash expense$12,402 52 %$7,446 23 %$4,956 67 %
Stock compensation10,965 46 %25,292 76 %(14,327)(57)%
Depreciation and amortization404 %403 %— %
Total general & administrative expense$23,771 100 %$33,141 100 %$(9,370)(28)%
Three Months Ended June 30, 2022% of
Expense
Category
Three Months Ended June 30, 2021% of
Expense
Category
Increase (Decrease)
$%
Salaries$3,175 10 %$2,570 14 %$605 24 %
Professional/outside services1,576 %379 %1,197 316 %
Facilities related703 %610 %93 15 %
Other G&A1,992 %2,278 12 %(286)(13)%
Total general & administrative expense, excluding non-cash expense$7,446 23 %$5,837 31 %$1,609 28 %
Stock compensation25,292 76 %12,020 66 %13,272 110 %
Depreciation/amortization403 %577 %(174)(30)%
Total general & administrative expense$33,141 100 %$18,434 100 %$14,707 80 %
Nine Months Ended June 30, 2022% of
Expense
Category
Nine Months Ended June 30, 2021% of
Expense
Category
Increase (Decrease)
$%
(in thousands)(in thousands)Nine Months Ended
June 30, 2023
% of
Expense
Category
Nine Months Ended
June 30, 2022
% of
Expense
Category
Increase (Decrease)
$%
SalariesSalaries$10,365 11 %$8,411 19 %$1,954 23 %Salaries$14,275 21 %$10,365 11 %$3,910 38 %
Professional/outside services6,083 %4,199 10 %1,884 45 %
Professional, outside services, and otherProfessional, outside services, and other15,293 22 %11,004 12 %4,289 39 %
Facilities relatedFacilities related2,085 %2,127 %(42)(2)%Facilities related3,377 %2,085 %1,292 62 %
Other G&A4,921 %4,326 10 %595 14 %
Total general & administrative expense, excluding non-cash expenseTotal general & administrative expense, excluding non-cash expense$23,454 25 %$19,063 44 %$4,391 23 %Total general & administrative expense, excluding non-cash expense$32,945 48 %$23,454 25 %$9,491 40 %
Stock compensationStock compensation67,739 74 %23,631 54 %44,108 187 %Stock compensation33,820 50 %67,739 73 %(33,919)(50)%
Depreciation/amortization1,210 %887 %323 36 %
Depreciation and amortizationDepreciation and amortization1,212 %1,210 %— %
Total general & administrative expenseTotal general & administrative expense$92,403 100 %$43,581 100 %$48,822 112 %Total general & administrative expense$67,977 100 %$92,403 100 %$(24,426)(26)%
Salaries expense increased by $605,000 from $2,570,000 during$1.9 million, or 59%, for the three months ended June 30, 2021 to $3,175,000 during the current period. Salaries expense increased by $1,954,000 from $8,411,000 during2023 and $3.9 million, or 38%, for the nine months ended June 30, 20212023 compared to $10,365,000 during the current period. Thissame period of 2022. The increase is primarily due to an increase in G&Awas driven by the combination of annual salary increases and increased headcount that has occurred as the Company has grown. We expect salaries expense to continue to increase as our headcount continues to increaserequired to support our expanding clinical pipeline, in addition to inflationary pressures in the labor market.Company’s growth.
Professional/Professional, outside services, includeand other expense includes legal, accounting, consulting, patent expenses, business insurance expenses, and other outside services, retained by the Company. Professional/outside servicestravel, communication and technology expenses. This expense increased by 1,197,000 from $379,000 during$2.4 million, or 68%, for the three months ended June 30, 2021 to $1,576,000 during the current period. Professional/outside services expense increased by $1,884,000 from $4,199,000 during2023 and $4.3 million, or 39%, for the nine months ended June 30, 20212023 compared to $6,083,000 during the current period. We expect future professional/outside services expense to increase as we continue to increase discovery efforts.same period of 2022. The increase in professional/outside services expense iswas mainly due to intellectual propertyconsulting expenses related to software implementation and general legal fees.administrative expenses in support of additional headcount.
28


Facilities-relatedFacilities related expense increased by $93,000 from $610,000 during the three months ended June 30, 2021 to $703,000 during the current period. Facilities-related expense decreased by $42,000 from $2,127,000 during the nine months ended June 30, 2021 to $2,085,000 during the current period. This category primarily includes rental costs and other facilities-related costs for ourthe Company’s corporate headquarters in Pasadena, California. The increase in facilitiesDepreciation and amortization expense, a noncash expense, was primarily related expenses is due to an increase in building maintenance and repair costs. We expect future facilities related expenses to increase as we continue to increase our headcount and footprint to support our discovery efforts.
Other G&A expense decreased by $286,000 from $2,278,000 duringamortization of leasehold improvements for the three months ended June 30, 2021 to $1,992,000 during the current period. Other G&A expense increased by $595,000 from $4,326,000 during the nine months ended June 30, 2021 to $4,921,000 during the current period. This category consists primarily of travel, communication and technology, office expenses, and franchise and property tax expenses. The decrease during the three months ended June 30, 2022 and 2021 is due to a decrease in consulting expenses from the prior year. The increase during the nine months ended June 30, 2022 and 2021 is due to software and software implementation costs..Company’s corporate headquarters.
Stock compensation expense, a non-cash expense, increaseddecreased by $13,272,000 from $12,020,000 during$14.3 million, or 57%, for the three months ended June 30, 2021 to $25,292,000 during the current period. Stock compensation expense, a non-cash expense, increased by $44,108,000 from $23,631,000 during2023 and $33.9 million, or 50%, for the nine months ended June 30, 20212023 compared to $67,739,000 during the current period. Stocksame periods of 2022. The decrease was mainly due to the lower amount of recognized compensation expense is based uponcosts and the valuationreversal of stock options and restricted stock units granted to employees, directors and certain consultants. Many variables affect the amount expensed, including the Company’s stock price on the date of the grant, as well as other assumptions. The increase in the current period is duerecognized compensation costs related to a performance award thatwhere the minimum performance goal was achieved earlier than anticipated, as well as a modification of certain performance awards to include market conditions.not met. The fair value of
32


market condition-based awards iswas expensed ratably over the service period and iswas not adjusted for actual achievement. We generally expect future
Other than with respect to the stock compensation expensecosts described above, the Company anticipates these general and administrative expenses to continue to increase as our headcount continuesits pipeline of candidates grows and progresses to increaselater phase clinical trials, in addition to support our clinical pipeline.
Depreciationinflationary pressure on goods and amortization expense, a noncash expense, decreased by $174,000 from $577,000 duringservices and the three months ended June 30, 2021 to $403,000 during the current period. Depreciation and amortization expense, a noncash expense, increased by $323,000 from $887,000 during the nine months ended June 30, 2021 to $1,210,000 during the current period. The decrease in depreciation expense in the current quarter is due to reclassification of certain depreciation from general and administrative to research and development.labor market.
Other Income (Loss)
Other income decreased by $1,080,000 from $1,944,000 during the three months ended June 30, 2021 to $863,000 during the current period. Other income decreased by $2,554,000 from $6,679,000 during the nine months ended June 30, 2021 compared to $4,125,000 during the current period. Other income(loss) is primarily related to interest income and realizedexpense. Other income decreased $1.5 million and unrealized gain/loss on our marketable securities.$5.0 million for the three and nine months ended June 30, 2023, respectively, compared to the same periods of 2022. The decrease in other income iswas primarily due to lower yieldsthe interest expense on more recently purchased bonds and increase in realized loss onthe liability related to the sale of marketable securitiesfuture royalties, offset by a property tax credithigher yields on investments due to increased interest rates as well as various credits the companyCompany received during the current period.first three quarters of fiscal 2023.
33

Liquidity and Capital Resources

ArrowheadLIQUIDITY AND CAPITAL RESOURCES
The Company has historically financed its operations through the sale of its equity securities, and revenue from its licensing and collaboration agreements.agreements, and the sale of certain future royalties. Research and development activities have required significant capital investment since the Company’s inception and are expected to continue to require significant cash expenditure as the Company’s pipeline continues to expand and matures into later stage clinical trials. Additionally, the Company plans to expandexpanded its facilities with its purchase of land in Verona, Wisconsin and its entry into a newcommenced the lease agreement for additional facilities in San Diego, California. Each of these expansions is designed to increase the Company’s internal manufacturing and discovery capabilities, and each will require significant capital investment.investment.
AtThe Company’s cash, cash equivalents and restricted cash decreased to $105.3 million at June 30, 2022, the Company's cash and cash equivalents totalled approximately $139.4 million as2023 compared to $184.4$108.0 million at September 30, 2021.2022. Cash invested in short-term fixed income securities and marketable securities was $277.1$346.4 million at June 30, 2022,2023 compared to $183.4$268.4 million at September 30, 2021.2022. Cash invested in long-term fixed income securities was $165.9$42.8 million at June 30, 2022,2023, compared to $245.6$105.9 million at September 30, 2021. The2022. On December 2, 2022, the Company also entered into anthe Open Market Sale Agreement, (the “ATM Agreement”) in August 2020, pursuant to which the Company may, from time to time, sell up to $250,000,000$250.0 million in shares of the Company’s Common Stockcommon stock through Jefferies LLC.LLC, acting as the sales agent and/or principal, in an at-the-market offering. As of June 30, 2022,2023, no shares have been soldissued under the ATMOpen Market Sale Agreement. The Company believes its current financial resources are sufficient to fund its operations through at least the next twelve months.
29


AThe following table presents a summary of cash flows forflows:
Nine Months Ended June 30,
20232022
(in thousands)
Cash Flow from:
Operating activities$(128,633)$(67,394)
Investing activities(126,664)(41,862)
Financing activities252,901 64,331 
Net decrease in cash, cash equivalents and restricted cash$(2,396)$(44,925)
Cash, cash equivalents and restricted cash at end of period$105,334 $139,439 
During the nine months ended June 30, 20222023, cash flows used by operating activities was $128.6 million, which was primarily due to the ongoing expenses related to the Company’s research and 2021 isdevelopment programs and general and administrative expenses, partially offset by the receipt of $110.0 million from collaboration and license agreements (see Note 2 — Collaboration and License Agreements to Consolidated Financial Statements of Part I, “Item 1. Financial Statements”). Cash used in investing activities was $126.7 million, which was primarily related to capital expenditures, $112.8 million of construction in progress, and $234.0 million purchases of investments, offset by maturities of investments of $220.2 million. Cash provided by financing activities of $252.9 million was primarily related to the $250.0 million payment from Royalty Pharma as follows:
Nine Months Ended June 30, 2022Nine Months Ended June 30, 2021
(in thousands)
Cash flow from:
Operating activities(67,464)195,341 
Investing activities(41,862)(23,433)
Financing activities64,331 10,490 
Net increase (decrease) in cash and cash equivalents(44,995)182,398 
Cash and cash equivalents at beginning of period184,434 143,583 
Cash and cash equivalents at end of period139,439 325,981 
well as cash received from stock option exercises.See Note 11 — Liability Related to the Sale of Future Royalties of Notes to Consolidated Financial Statements of Part I, “Item 1. Financial Statements.”
During the nine months ended June 30, 2022, cash flowflows used by operating activities was $67.5$67.4 million, which was primarily due to the receipt of the $120.0 million upfront payment from GSK, offset by the ongoing expenses related to the Company’s research and development programs and general and administrative expenses. Cash used in investing activities was $41.9 million, which was primarily related to the purchase of property and equipment of $20.1 million and net salespurchases and maturities of investments of $21.8 million. Cash provided by financing activities of $64.3 million was primarily related to the formation of ourthe Company's joint venture, Viserna,Visirna, as well as cash received from stock option exercises.
During the nine months ended June 30, 2021, cash flow provided by operating activities was $195.3 million, which was primarily due to the $300 million payment received under the Takeda License Agreement, partially offset by ongoing expenses of the Company’s research and development programs and general and administrative expenses. Cash used in investing activities was $23.4 million, which was primarily related to the purchase of property and equipment of $15.4 million, partially offset by the net sales of investments of $8.1 million. Cash provided by financing activities of $10.5 million was related to cash received from stock option exercises.
On December 20, 2021, the Company completed a purchase of 13 acres of land in the Verona Technology Park in Verona, Wisconsin, which is planned to be the site ofbeing developed into an approximately 160,000 square foot drug manufacturing facility and an approximately 140,000 square foot laboratory and office facility towhich will support the Company’s process development and analytical activities. ArrowheadThe Company has incurred $102.7 million and intends to invest between $200spend an additional $160.0 million and $250to $180.0 million intoto complete the buildoutbuild out of the facilities.facilities with cash on hand. As part of this acquisition,land purchase, the Company also entered into a development agreement with the City of Verona to construct certain infrastructure improvements within the TIFtax incremental district and willexpects to be reimbursed up to $16.0 million by the City of Verona by future tax increment revenue generated from the developed property. The total amount of funding that City of Verona willis expected to pay as reimbursements under the TIFTax Incremental Financing program for these improvements is not guaranteed and will depend on future tax revenues generated from the developed property. The Company also expects receive up to $2.5 million of refundable Wisconsin state income tax
Capital Resources
34


credits from the Wisconsin Economic Development Corporation (WEDC) as incentives to invest in the local community and Material Cash Requirements
A summary of our capital resources and material cash requirements is presented in Item 7 of our Annual Report on Form 10-K for our fiscal year ended September 30, 2021. Other than as described above, there were no material changes to our capital resources and material cash requirements during the three months ended June 30, 2022.create new jobs.
ITEM 3.    QUANTITATIVE AND QUALITATIVE DISCLOSURES ABOUT MARKET RISK
There has been no material change in ourthe Companys exposure to market risk from that described in Item 7A of ourits Annual Report on Form 10-K for the year ended September 30, 2021.2022.
ITEM 4.    CONTROLS AND PROCEDURES
Our Chief Executive OfficerEvaluation of Disclosure Controls and our Chief Financial Officer, after evaluating our “disclosure controls and procedures” (as defined in Rules 13a-15(e) and 15d-15(e) of the Securities Exchange Act of 1934, as amended (the “Exchange Act”)) as of the end of the period covered by this Quarterly Report on Form 10-Q (the “Evaluation Date”), have concluded that, as of the Evaluation Date, ourProcedures
The Company maintains disclosure controls and procedures are effectivedesigned to ensure that information we are required to disclosebe disclosed in its reports that we file or submitfiled under the Exchange Act is recorded, processed, summarized,
30


and reported within the time periods specified in Securities and Exchange Commissionthe SEC rules and forms, and to ensure that information required to be disclosed by us in such reportsinformation is accumulated and communicated to ourits management, including ourits Chief Executive Officer and Chief Financial Officer, whereas appropriate, to allow for timely decisions regarding required disclosure. In designing and evaluating the disclosure controls and procedures, management recognizes that any controls and procedures, no matter how well designed and operated, can provide only reasonable assurance of achieving the desired control objectives.
NoAs required by Rule 13a-15(b) of the Exchange Act, the Company carried out an evaluation, under the supervision and with the participation of its management, including its Chief Executive Officer and Chief Financial Officer, of the effectiveness of the design and operation of the Company’s disclosure controls and procedures as of the end of the quarter covered by this Quarterly Report on Form 10-Q. Based on the foregoing, the Company’s Chief Executive Officer and Chief Financial Officer concluded that the Company’s disclosure controls and procedures were effective at the reasonable assurance level.
Changes in Internal Control Over Financial Reporting
There has been no change in the Company’s internal controlscontrol over financial reporting (as defined in Rules 13a-15(f) and 15d-15(f) of the Exchange Act) occurred during the Company’s most recent fiscal quarter that has materially affected, or is reasonably likely to materially affect, ourthe Company’s internal control over financial reporting. The Company regularly evaluates its controls and procedures and makes improvements in the design and effectiveness of established controls and procedures and the remediation of any deficiencies which may be identified during this process.


3135


PART II—OTHER INFORMATION
ITEM 1.    LEGAL PROCEEDINGS
From time to time, wethe Company may be involved in routine legal proceedings, as well as demands, claims and threatened litigation, which arise in the normal course of ourits business. Litigation can be expensive and disruptive to normal business operations. Moreover, the results of legal proceedings, particularly complex legal proceedings, cannot be predicted with any certainty. There have been no material developments in the legal proceedings that wethe Company disclosed in Part I, Item 3 of ourits Annual Report on Form 10-K for the year ended September 30, 2021.2022.
ITEM 1A.    Risk FactorsRISK FACTORS

The Company
There have been no material changess business, results of operations and financial conditions are subject to various risks. These risks are described elsewhere in this Quarterly Report on Form 10-Q and in the risk factors included in ourCompanys other filings with the SEC, including the Companys Annual Report on Form 10-K for the year ended September 30, 2021 as updated by our Quarterly Report on Form 10-Q for the period ended March 31, 2022. Please carefully consider the information set forth in this Quarterly Report on Form 10-Q andThere have been no material changes from the risk factors discussedidentified in Part I, “Item 1A. Risk Factors” in ourthe Companys Annual Report on Form 10-K for the year ended September 30, 2021 and in Part II, “Item 1A. Risk Factors” in our Quarterly Report on Form 10-Q for the period ended March 31, 2022, which could materially affect our business, financial condition or future results. The risks described in our Annual Report on Form 10-K and our Quarterly Report on Form 10-Q, as well as other risks and uncertainties, could materially and adversely affect our business, results of operations and financial condition, which in turn could materially and adversely affect the trading price of shares of our Common Stock. Additional risks not currently known or currently material to us may also harm our business.2022.
ITEM 2.    UNREGISTERED SALES OF EQUITY SECURITIES AND USE OF PROCEEDS
None.
ITEM 3.    DEFAULTS UPON SENIOR SECURITIES
None.
ITEM 4.    MINE SAFETY DISCLOSURES
Not Applicable.
ITEM 5.    OTHER INFORMATION
(c) Trading Plans
During the quarter ended June 30, 2023, no director or officer adopted or terminated any Rule 10b5-1 trading arrangement or non-Rule 10b5-1 trading arrangement.

None.
3236


ITEM 6.    EXHIBITS
Exhibit
Number
Document Description
3.1
3.2
3.3
Second Amended and Restated Bylaws of Arrowhead Pharmaceuticals, Inc., as amended January 24, 2023 (incorporated by reference from Exhibit 3.3 of the Company’s Form 10-Q filed on May 2, 2023)
31.1*
31.2*
32.1**
32.2**
101.INS*Inline XBRL Instance Document
101.SCH*Inline XBRL Taxonomy Extension Schema Document
101.CAL*Inline XBRL Taxonomy Extension Calculation Linkbase Document
101.LAB*Inline XBRL Taxonomy Extension Label Linkbase Document
101.PRE*Inline XBRL Taxonomy Extension Presentation Linkbase Document
101.DEF*Inline XBRL Taxonomy Extension Definition Linkbase Document
104*The cover page from this Quarterly Report on Form 10-Q, formatted in Inline XBRL (included as Exhibit 101)
_________________
*Filed herewith.
**Furnished herewith.

3337


SIGNATURE
Pursuant to the requirements of the Securities Exchange Act of 1934, the registrant has duly caused this Quarterly Report on Form 10-Q to be signed on its behalf by the undersigned, thereunto duly authorized.
Dated: August 4, 20227, 2023
ARROWHEAD PHARMACEUTICALS, INC.
By:/s/ Kenneth A. Myszkowski
Kenneth A. Myszkowski
Chief Financial Officer
(Principal Financial Officer and Duly Authorized Officer)
3438