FYfalse--12-310001761918April 30,
20223May 31,
20240001761918us-gaap:CommonStockMembereras:ErascaFoundationMember2021-01-012021-12-310001761918us-gaap:RestrictedStockMember2020-01-012020-12-310001761918us-gaap:CommonStockMember2019-12-310001761918eras:TheRegentsOfUniversityOfCaliforniaMembereras:LicenseAgreementMember2021-08-012021-08-310001761918srt:MaximumMembereras:LicenseAgreementMembereras:LifeArcMember2020-04-012020-04-300001761918srt:MaximumMemberus-gaap:EmployeeStockOptionMember2020-01-012020-12-310001761918us-gaap:MoneyMarketFundsMemberus-gaap:CashAndCashEquivalentsMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:CommonStockMember2020-12-310001761918us-gaap:RetainedEarningsMember2021-12-310001761918srt:MaximumMemberus-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMember2020-12-310001761918us-gaap:AdditionalPaidInCapitalMember2020-12-310001761918us-gaap:ShortTermInvestmentsMembereras:SupranationalDebtSecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918eras:SeriesB2ConvertiblePreferredStockMemberus-gaap:CommonStockMemberus-gaap:IPOMembereras:AsanaMergerAgreementMember2020-11-012020-11-300001761918us-gaap:CommonStockMembereras:ErascaFoundationMember2021-07-012021-07-310001761918us-gaap:FairValueInputsLevel1Memberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918eras:LongTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2021-12-3100017619182021-01-012021-12-310001761918us-gaap:AccountingStandardsUpdate201815Member2021-12-310001761918eras:SecondLicensedProductMembereras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2020-02-012020-02-290001761918us-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:RetainedEarningsMember2021-01-012021-12-310001761918srt:MaximumMembereras:LicenseAgreementMembereras:KatmaiPharmaceuticalsIncorporationMember2020-03-012020-03-310001761918us-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:AdditionalPaidInCapitalMemberus-gaap:IPOMember2021-01-012021-12-310001761918us-gaap:ConvertiblePreferredStockMember2019-12-310001761918us-gaap:EmployeeStockOptionMember2021-12-3100017619182021-12-310001761918srt:MinimumMembereras:EmployeeStockPurchasePlanMember2021-07-012021-07-310001761918eras:LicenseAgreementMembereras:KatmaiPharmaceuticalsIncorporationMember2021-01-012021-12-310001761918us-gaap:EmployeeStockOptionMember2020-01-012020-12-310001761918us-gaap:CommonStockMember2021-12-310001761918us-gaap:AdditionalPaidInCapitalMember2021-01-012021-12-310001761918eras:LicenseAgreementMembereras:LifeArcMember2020-01-012020-12-310001761918eras:SeriesB2ConvertiblePreferredStockMember2021-01-310001761918eras:SharesAvailableForPurchaseUnderEmployeeStockPurchasePlanMember2021-12-310001761918us-gaap:LeaseholdImprovementsMember2021-12-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:CommercialPaperMemberus-gaap:CashAndCashEquivalentsMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:ResearchAndDevelopmentExpenseMember2021-01-012021-12-310001761918us-gaap:CorporateDebtSecuritiesMembersrt:MaximumMemberus-gaap:ShortTermInvestmentsMember2020-12-310001761918eras:ErascaFoundationMember2021-01-012021-12-3100017619182020-01-012020-12-310001761918us-gaap:RetainedEarningsMember2019-12-310001761918eras:SeriesAConvertiblePreferredStockMember2019-12-310001761918eras:AsanaMergerAgreementMember2020-12-310001761918eras:TwoThousandEighteenLeaseMember2021-11-012021-11-300001761918us-gaap:AccumulatedOtherComprehensiveIncomeMember2019-12-310001761918us-gaap:DomesticCountryMemberus-gaap:ResearchMember2021-01-012021-12-310001761918us-gaap:CommonStockMemberus-gaap:IPOMember2021-01-012021-12-310001761918us-gaap:RestrictedStockMember2021-01-012021-12-310001761918eras:LicenseAgreementMembereras:LifeArcMember2021-01-012021-12-310001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2020-04-012020-04-300001761918us-gaap:AdditionalPaidInCapitalMember2021-12-310001761918us-gaap:CommonStockMemberus-gaap:IPOMember2021-07-200001761918srt:MinimumMemberus-gaap:EmployeeStockOptionMember2020-01-012020-12-310001761918eras:OtherStateMember2021-12-310001761918eras:SeriesB1ConvertiblePreferredStockMember2020-12-310001761918eras:TwoThousandTwentyLeaseMember2020-09-012020-09-300001761918us-gaap:ConvertiblePreferredStockMember2020-01-012020-12-310001761918eras:SupranationalDebtSecuritiesMembersrt:MaximumMemberus-gaap:ShortTermInvestmentsMember2021-12-310001761918eras:TwoThousandTwentyLeaseMember2020-09-300001761918srt:MinimumMember2021-01-012021-12-310001761918eras:EmployeeStockPurchasePlanMember2021-07-310001761918us-gaap:AccountsPayableAndAccruedLiabilitiesMember2020-12-310001761918eras:AsanaMergerAgreementMember2020-11-300001761918eras:PreferredStockPurchaseRightLiabilityMember2021-01-012021-12-310001761918us-gaap:CommonStockMember2020-01-012020-12-310001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2020-01-012020-12-310001761918srt:MinimumMembereras:AtTimeOptionIsGrantedMemberus-gaap:EmployeeStockOptionMember2021-01-012021-12-310001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-3100017619182021-07-200001761918us-gaap:ShareBasedCompensationAwardTrancheTwoMemberus-gaap:RestrictedStockMember2018-01-012018-12-310001761918eras:TheRegentsOfUniversityOfCaliforniaMembereras:LicenseAgreementMemberus-gaap:CommonStockMember2021-05-012021-05-310001761918us-gaap:RestrictedStockMember2018-01-012018-12-310001761918eras:MeasurementInputFairValueOfUnderlyingPreferredStockMembereras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMember2021-01-200001761918us-gaap:RestrictedStockMemberus-gaap:ShareBasedCompensationAwardTrancheOneMember2021-01-012021-12-310001761918eras:AwardsAvailableForFutureGrantMember2021-12-310001761918eras:TheRegentsOfUniversityOfCaliforniaMembereras:LicenseAgreementMember2020-01-012020-12-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:ShortTermInvestmentsMemberus-gaap:CorporateDebtSecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918eras:TheRegentsOfUniversityOfCaliforniaMembersrt:MaximumMembereras:LicenseAgreementMember2021-05-012021-05-310001761918eras:SeriesB2ConvertiblePreferredStockMember2020-12-3100017619182020-04-150001761918eras:TwoThousandTwentyOneLeaseMember2021-01-012021-12-310001761918us-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918eras:TwoThousandTwentyOneIncentiveAwardPlanMember2021-07-012021-07-310001761918us-gaap:CommonStockMembereras:EmergeLifeSciencesPurchaseAgreementMember2021-03-012021-03-310001761918eras:ConversionOfB2PreferredStockInFutureIssuanceMember2020-12-310001761918us-gaap:RestrictedStockMember2018-12-310001761918us-gaap:EmployeeStockOptionMember2021-01-012021-12-310001761918us-gaap:IPOMember2021-07-200001761918us-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2021-12-310001761918us-gaap:DomesticCountryMember2021-12-310001761918us-gaap:CommonStockMember2021-01-012021-12-310001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2020-12-310001761918us-gaap:DomesticCountryMember2021-01-012021-12-310001761918eras:FirstAmendmentToTwoThousandAndTwentyLeaseMemberus-gaap:LetterOfCreditMember2021-03-3100017619182022-03-170001761918eras:EmployeeStockPurchasePlanMember2020-01-012020-12-310001761918us-gaap:OfficeEquipmentMember2021-12-310001761918us-gaap:MeasurementInputRiskFreeInterestRateMembereras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMember2020-12-310001761918eras:EmergeLifeSciencesPurchaseAgreementMember2020-01-012020-12-310001761918us-gaap:OtherNonoperatingIncomeExpenseMember2021-01-012021-12-310001761918us-gaap:PrepaidExpensesAndOtherCurrentAssetsMembereras:ErascaFoundationMember2021-12-310001761918us-gaap:RestrictedStockMember2021-12-310001761918eras:AsanaMergerAgreementMember2021-01-012021-12-310001761918eras:TwoThousandEighteenLeaseMember2021-10-312021-10-310001761918us-gaap:IPOMember2021-01-012021-12-310001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMemberus-gaap:MeasurementInputPriceVolatilityMember2021-01-200001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2020-02-012020-02-290001761918eras:SeriesB2ConvertiblePreferredStockMembereras:AsanaMergerAgreementMember2020-11-300001761918us-gaap:CaliforniaFranchiseTaxBoardMemberus-gaap:ResearchMember2021-12-310001761918eras:OptionsEarlyExercisedSubjectToFutureVestingMember2021-01-012021-12-310001761918us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-12-310001761918eras:EstimatedSharesPurchasableUnderTheEmployeeStockPurchasePlanMember2021-01-012021-12-310001761918eras:SeriesB2ConvertiblePreferredStockMembereras:LicenseAgreementMembereras:KatmaiPharmaceuticalsIncorporationMember2021-01-012021-01-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:ShortTermInvestmentsMemberus-gaap:CorporateDebtSecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:ComputerEquipmentMember2020-12-310001761918eras:LicenseAgreementMembereras:LifeArcMember2020-04-012020-04-300001761918eras:AwardsAvailableForFutureGrantMember2020-12-310001761918us-gaap:ConvertiblePreferredStockMember2020-12-310001761918srt:MaximumMemberus-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMember2021-12-310001761918us-gaap:FairValueInputsLevel1Memberus-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:SoftwareAndSoftwareDevelopmentCostsMember2020-12-310001761918srt:MaximumMembereras:LongTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2021-12-310001761918eras:TwoThousandEighteenLeaseMember2018-12-3100017619182021-08-012021-08-310001761918us-gaap:EmployeeStockOptionMember2020-01-012020-12-310001761918srt:MaximumMembereras:LicenseAgreementMembereras:LifeArcMember2020-03-012020-03-310001761918eras:TwoThousandEighteenEquityIncentivePlanMember2021-08-012021-12-310001761918srt:MinimumMembereras:EmployeeStockPurchasePlanMember2021-01-012021-12-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:ShortTermInvestmentsMembereras:SupranationalDebtSecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:OverAllotmentOptionMemberus-gaap:CommonStockMember2021-07-202021-07-200001761918eras:AsanaMergerAgreementMember2020-11-012020-11-300001761918us-gaap:CorporateDebtSecuritiesMemberus-gaap:ShortTermInvestmentsMember2021-12-310001761918eras:ErascaFoundationMember2021-12-012021-12-310001761918us-gaap:DomesticCountryMemberus-gaap:ResearchMember2021-12-310001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMemberus-gaap:MeasurementInputPriceVolatilityMember2020-12-310001761918srt:MaximumMemberus-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2020-12-310001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2021-01-012021-12-310001761918eras:LicenseAgreementMembereras:SeriesB1ConvertiblePreferredStockMembereras:KatmaiPharmaceuticalsIncorporationMember2020-04-012020-04-300001761918us-gaap:ShortTermInvestmentsMemberus-gaap:CorporateDebtSecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918eras:LicenseAgreementMembereras:KatmaiPharmaceuticalsIncorporationMember2020-01-012020-12-310001761918eras:RestrictedStockSubjectToFutureVestingMember2020-01-012020-12-310001761918eras:SeriesB2ConvertiblePreferredStockMember2021-01-012021-01-310001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMemberus-gaap:MeasurementInputExpectedDividendRateMember2021-01-200001761918us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-01-012020-12-310001761918eras:OptionsEarlyExercisedSubjectToFutureVestingMember2020-01-012020-12-310001761918eras:LicenseAgreementMembereras:SeriesB1ConvertiblePreferredStockAndSeriesB2ConvertiblePreferredStockMembereras:KatmaiPharmaceuticalsIncorporationMember2020-03-012020-03-310001761918eras:TwoThousandTwentyOneLeaseMember2021-12-012021-12-310001761918us-gaap:GeneralAndAdministrativeExpenseMember2021-01-012021-12-310001761918us-gaap:FairValueInputsLevel1Memberus-gaap:MoneyMarketFundsMemberus-gaap:CashAndCashEquivalentsMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-01-012021-12-310001761918us-gaap:ConvertiblePreferredStockMember2021-12-310001761918eras:SeriesAConvertiblePreferredStockMember2018-01-012019-12-310001761918eras:SeriesBConvertiblePreferredStockPurchaseAgreementMembereras:SeriesB1ConvertiblePreferredStockMember2020-04-300001761918srt:MaximumMember2021-01-012021-12-310001761918eras:ForHoldersOfMoreThanTenPercentOfCombinedVotingPowerOfAllClassesOfStockMembersrt:MinimumMemberus-gaap:EmployeeStockOptionMember2021-01-012021-12-310001761918us-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:AdditionalPaidInCapitalMembereras:ErascaFoundationMember2021-01-012021-12-3100017619182021-02-280001761918eras:SharesAvailableForPurchaseUnderEmployeeStockPurchasePlanMember2020-12-310001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:MeasurementInputExpectedTermMemberus-gaap:ValuationTechniqueOptionPricingModelMember2021-01-202021-01-200001761918eras:EmployeeStockPurchasePlanMember2021-12-310001761918eras:TheRegentsOfUniversityOfCaliforniaMembereras:LicenseAgreementMember2021-05-012021-05-3100017619182020-12-310001761918us-gaap:AccountsPayableAndAccruedLiabilitiesMember2021-12-310001761918eras:SeriesAConvertiblePreferredStockMember2020-12-310001761918us-gaap:OfficeEquipmentMember2020-12-310001761918eras:SeriesB2ConvertiblePreferredStockMembereras:AsanaMergerAgreementMember2020-11-012020-11-300001761918us-gaap:ConvertiblePreferredStockMember2021-01-012021-12-310001761918eras:PerformanceBasedOptionsMember2021-01-012021-12-310001761918eras:TwoThousandTwentyOneIncentiveAwardPlanMember2021-07-310001761918us-gaap:ConstructionInProgressMember2021-12-310001761918eras:EmployeeStockPurchasePlanMember2021-07-012021-07-310001761918eras:LongTermInvestmentsMemberus-gaap:USTreasurySecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918eras:SeriesAConvertiblePreferredStockMember2018-12-3100017619182019-12-310001761918srt:MaximumMemberus-gaap:EmployeeStockOptionMember2021-01-012021-12-310001761918us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-12-310001761918us-gaap:FurnitureAndFixturesMember2020-12-310001761918us-gaap:EmployeeStockOptionMember2021-01-012021-12-310001761918eras:AsanaMergerAgreementMember2020-01-012020-12-310001761918us-gaap:MeasurementInputRiskFreeInterestRateMembereras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMember2021-01-200001761918eras:SupranationalDebtSecuritiesMemberus-gaap:ShortTermInvestmentsMember2021-12-310001761918eras:TheRegentsOfUniversityOfCaliforniaMembereras:LicenseAgreementMember2021-01-012021-12-310001761918eras:SeriesB2ConvertiblePreferredStockMember2021-12-310001761918eras:PerformanceBasedOptionsMember2020-01-012020-12-310001761918eras:TwoThousandTwentyOneLeaseMember2021-12-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918eras:SeriesBConvertiblePreferredStockPurchaseAgreementMembereras:SeriesB1ConvertiblePreferredStockMember2020-04-012020-04-300001761918us-gaap:RetainedEarningsMember2020-12-310001761918eras:RestrictedStockSubjectToFutureVestingMember2021-01-012021-12-310001761918us-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918eras:EmergeLifeSciencesPurchaseAgreementMember2021-01-012021-12-310001761918us-gaap:GeneralAndAdministrativeExpenseMember2020-01-012020-12-310001761918eras:LaboratoryEquipmentMember2021-12-310001761918srt:MaximumMemberus-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2021-12-310001761918eras:LicenseAgreementMembereras:KatmaiPharmaceuticalsIncorporationMember2020-03-012020-03-310001761918us-gaap:RestrictedStockMemberus-gaap:ShareBasedCompensationAwardTrancheOneMember2018-01-012018-12-310001761918us-gaap:CaliforniaFranchiseTaxBoardMember2021-01-012021-12-310001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2020-02-290001761918eras:ConversionOfB2PreferredStockInFutureIssuanceMember2021-12-310001761918srt:MinimumMembereras:LongTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2021-12-310001761918us-gaap:ResearchMemberstpr:MA2021-01-012021-12-310001761918us-gaap:ResearchMemberstpr:MA2021-12-310001761918us-gaap:FairValueInputsLevel1Membereras:LongTermInvestmentsMemberus-gaap:USTreasurySecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:FairValueInputsLevel1Memberus-gaap:CashAndCashEquivalentsMemberus-gaap:MoneyMarketFundsMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:CaliforniaFranchiseTaxBoardMember2021-12-310001761918srt:MaximumMembereras:EmployeeStockPurchasePlanMember2021-01-012021-12-310001761918us-gaap:ComputerEquipmentMember2021-12-310001761918eras:MeasurementInputFairValueOfUnderlyingPreferredStockMembereras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMember2020-12-310001761918us-gaap:RestrictedStockMemberus-gaap:ShareBasedCompensationAwardTrancheThreeMember2018-01-012018-12-310001761918srt:MaximumMembereras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMembereras:FirstLicensedProductMember2020-02-012020-02-290001761918us-gaap:FairValueInputsLevel1Memberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918stpr:CAeras:TwoThousandEighteenLeaseMember2018-12-310001761918eras:EmergeLifeSciencesPurchaseAgreementMember2021-03-012021-03-310001761918us-gaap:LeaseholdImprovementsMember2020-12-310001761918us-gaap:AdditionalPaidInCapitalMember2019-12-310001761918eras:PreferredStockPurchaseRightLiabilityMember2020-01-012020-12-310001761918eras:NiKangTherapeuticsIncorporationMembereras:LicenseAgreementMember2021-12-310001761918eras:SeriesB2ConvertiblePreferredStockMember2020-01-012020-12-310001761918us-gaap:SoftwareAndSoftwareDevelopmentCostsMember2021-12-310001761918us-gaap:RestrictedStockMember2020-12-310001761918us-gaap:ShortTermInvestmentsMemberus-gaap:CorporateDebtSecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918eras:FirstAmendmentToTwoThousandAndTwentyLeaseMember2021-03-310001761918eras:FirstAmendmentToTwoThousandAndTwentyLeaseMember2021-03-012021-03-310001761918us-gaap:IPOMember2021-02-280001761918eras:AsanaMergerAgreementMember2021-12-310001761918eras:LaboratoryEquipmentMember2020-12-310001761918us-gaap:CommonStockMemberus-gaap:IPOMember2021-07-012021-07-310001761918us-gaap:CorporateDebtSecuritiesMemberus-gaap:ShortTermInvestmentsMember2020-12-310001761918us-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMember2020-12-310001761918eras:AffiniTTherapeuticsIncMembersrt:SubsidiariesMemberus-gaap:SubsequentEventMember2022-03-240001761918us-gaap:CommonStockMembereras:EmergeLifeSciencesPurchaseAgreementMember2021-03-310001761918us-gaap:OtherNoncurrentAssetsMembereras:TwoThousandTwentyOneLeaseMember2021-12-310001761918eras:SeriesBConvertiblePreferredStockPurchaseAgreementMember2020-04-012020-04-300001761918us-gaap:MoneyMarketFundsMemberus-gaap:CashAndCashEquivalentsMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMember2020-12-310001761918us-gaap:AdditionalPaidInCapitalMember2020-01-012020-12-310001761918us-gaap:CommonStockMemberus-gaap:IPOMember2021-07-202021-07-200001761918us-gaap:FairValueInputsLevel1Memberus-gaap:ShortTermInvestmentsMemberus-gaap:USTreasurySecuritiesMemberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:LetterOfCreditMembereras:TwoThousandTwentyLeaseMember2020-09-012020-09-300001761918us-gaap:FairValueInputsLevel2Memberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918eras:OtherStateMember2021-01-012021-12-310001761918eras:LicenseAgreementMembereras:SeriesB1ConvertiblePreferredStockAndSeriesB2ConvertiblePreferredStockMemberus-gaap:IPOMembereras:KatmaiPharmaceuticalsIncorporationMember2020-03-310001761918us-gaap:CommercialPaperMemberus-gaap:CashAndCashEquivalentsMemberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:FairValueInputsLevel3Memberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918us-gaap:FurnitureAndFixturesMember2021-12-310001761918us-gaap:CommercialPaperMemberus-gaap:ShortTermInvestmentsMember2021-12-310001761918us-gaap:LetterOfCreditMembereras:TwoThousandTwentyLeaseMember2020-09-3000017619182021-07-092021-07-090001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:ValuationTechniqueOptionPricingModelMemberus-gaap:MeasurementInputExpectedDividendRateMember2020-12-310001761918us-gaap:ResearchAndDevelopmentExpenseMember2020-01-012020-12-310001761918eras:EmployeeStockPurchasePlanMember2021-01-012021-12-310001761918us-gaap:EmployeeStockOptionMember2021-01-012021-01-310001761918eras:ConversionOfB2PreferredStockInFutureIssuanceMember2020-01-012020-12-310001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:FairValueInputsLevel3Memberus-gaap:FairValueMeasurementsRecurringMember2020-12-310001761918eras:SeriesB2ConvertiblePreferredStockMembereras:SeriesBConvertiblePreferredStockPurchaseAgreementMember2020-04-300001761918eras:PreferredStockPurchaseRightLiabilityMemberus-gaap:MeasurementInputExpectedTermMemberus-gaap:ValuationTechniqueOptionPricingModelMember2020-01-012020-12-310001761918us-gaap:FairValueInputsLevel2Memberus-gaap:FairValueMeasurementsRecurringMember2021-12-310001761918us-gaap:EmployeeStockOptionMember2020-12-310001761918us-gaap:RetainedEarningsMember2020-01-012020-12-310001761918us-gaap:CorporateDebtSecuritiesMembersrt:MaximumMemberus-gaap:ShortTermInvestmentsMember2021-12-310001761918eras:TwoThousandTwentyOneIncentiveAwardPlanMember2021-12-31eras:Litigationeras:Positioneras:Segmentxbrli:pureutr:sqftxbrli:sharesiso4217:USDxbrli:sharesiso4217:USD
UNITED STATES
SECURITIES AND EXCHANGE COMMISSION
Washington, D.C. 20549
FORM
10-K
(Mark One)
|
|
☒
|
ANNUAL REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES
EXCHANGE ACT OF 1934
|
For the fiscal year ended
DECEMBER 31,
2021
OR
|
|
☐
|
TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES
EXCHANGE ACT OF 1934 FOR THE TRANSITION PERIOD FROM TO
|
Commission File Number
001-40602
ERASCA, INC.
(Exact name of Registrant as specified in its Charter)
|
|
Delaware
|
83-1217027
|
(State or other jurisdiction of
incorporation or organization)
|
(I.R.S. Employer
Identification No.)
|
10835 Road to the Cure,
Suite 140
San Diego,
CA
|
92121
|
(Address of principal executive offices)
|
(Zip Code)
|
Registrant’s telephone number, including area code:
(858)
465-6511
Securities registered pursuant to Section 12(b) of the
Act:
|
|
|
|
|
Title of each class
|
|
Trading
Symbol(s)
|
|
Name of each exchange on which registered
|
Common Stock, $0.0001 par value per share
|
|
ERAS
|
|
Nasdaq Global Select Market
|
Securities registered pursuant to Section 12(g) of the Act:
None
Indicate by check mark if the Registrant is a well-known seasoned
issuer, as defined in Rule 405 of the Securities Act. YES
☐No
☒
Indicate by check mark if the Registrant is not required to file
reports pursuant to Section 13 or 15(d) of the Act. YES ☐
No
☒
Indicate by check mark whether the Registrant: (1) has filed all
reports required to be filed by Section 13 or 15(d) of the
Securities Exchange Act of 1934 during the preceding 12 months (or
for such shorter period that the Registrant was required to file
such reports), and (2) has been subject to such filing requirements
for the past 90 days.
Yes
☒
NO
☐
Indicate by check mark whether the Registrant has submitted
electronically every Interactive Data File required to be submitted
pursuant to Rule 405 of Regulation S-T (§232.405 of this chapter)
during the preceding 12 months (or for such shorter period that the
Registrant was required to submit such files).
Yes
☒NO
☐
Indicate by check mark whether the registrant is a large
accelerated filer, an accelerated filer, a non-accelerated filer,
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 filer
|
|
☐
|
|
Accelerated filer
|
|
☐
|
|
|
|
|
Non-accelerated filer
|
|
☒
|
|
Smaller reporting company
|
|
☒
|
|
|
|
|
|
|
|
Emerging growth company
|
|
☒
|
|
|
|
|
If an emerging growth company, indicate by check mark if the
registrant has elected not to use the extended transition period
for complying with any new or revised financial accounting
standards provided pursuant to Section 13(a) of the Exchange
Act.
☐
Indicate by check mark whether the registrant has filed a report on
and attestation to its management’s assessment of the effectiveness
of its internal control over financial reporting under Section
404(b) of the Sarbanes-Oxley Act (15 U.S.C. 7262(b)) by the
registered public accounting firm that prepared or issued its audit
report.
☐
Indicate by check mark whether the Registrant is a shell company
(as defined in Rule 12b-2 of the Exchange Act). YES ☐ NO
☒
The aggregate market value of the Registrant's common stock held by
non-affiliates of the Registrant was
approximately $1.1
billion as
of the closing of the Registrant’s initial public offering on July
20, 2021 (based on the closing price of $19.23 per share on the
Nasdaq Global Select Market as of such date).
The number of shares of Registrant’s Common Stock outstanding as of
March 17, 2022 was
121,592,318.
DOCUMENTS INCORPORATED BY REFERENCE
Certain sections of the Registrant’s definitive proxy statement for
the 2022 annual meeting of stockholders to be filed with the
Securities and Exchange Commission pursuant to Regulation 14A not
later than 120 days after the end of the fiscal year covered by
this Form 10-K are incorporated by reference into Part III of this
Form 10-K.
Table of Contents
i
PART
I
Forward-Looking Statements and Market Data
This Annual Report on Form 10-K contains forward-looking statements
within the meaning of Section 21E of the Securities Exchange Act of
1934, as amended (the Exchange Act), and Section 27A of the
Securities Act of 1933, as amended (the Securities Act). All
statements other than statements of historical facts contained in
this Annual Report on Form 10-K, including statements regarding our
future results of operations and financial position, business
strategy, research and development plans, the anticipated timing,
costs, design and conduct of our ongoing and planned preclinical
studies and planned clinical trials for our product candidates, the
timing and likelihood of regulatory filings and approvals for our
product candidates, our ability to commercialize our product
candidates, if approved, the impact of the COVID-19 pandemic on our
business, the pricing and reimbursement of our product candidates,
if approved, the potential to develop future product candidates,
the potential benefits of strategic agreements and our intent to
enter into any strategic arrangements, the timing and likelihood of
success, plans and objectives of management for future operations,
and future results of anticipated product development efforts, are
forward-looking statements. These statements involve known and
unknown risks, uncertainties and other important factors that may
cause our actual results, performance or achievements to be
materially different from any future results, performance or
achievements expressed or implied by the forward-looking
statements. This Annual Report on Form 10-K also contains estimates
and other statistical data made by independent parties and by us
relating to market size and growth and other data about our
industry. This data involves a number of assumptions and
limitations, and you are cautioned not to give undue weight to such
estimates. In addition, projections, assumptions and estimates of
our future performance and the future performance of the markets in
which we operate are necessarily subject to a high degree of
uncertainty and risk.
In some cases, you can identify forward-looking statements by terms
such as “may,” “will,” “should,” “expect,” “plan,” “anticipate,”
“could,” “intend,” “target,” “project,” “contemplates,” “believes,”
“estimates,” “predicts,” “potential” or “continue” or the negative
of these terms or other similar expressions. The forward-looking
statements in this Annual Report on Form 10-K are only predictions.
We have based these forward-looking statements largely on our
current expectations and projections about future events and
financial trends that we believe may affect our business, financial
condition and results of operations. These forward-looking
statements speak only as of the date of this Annual Report on Form
10-K and are subject to a number of risks, uncertainties and
assumptions, including those described in Part I, Item 1A, “Risk
Factors.” The events and circumstances reflected in our
forward-looking statements may not be achieved or occur, and actual
results could differ materially from those projected in the
forward-looking statements. Moreover, we operate in an evolving
environment. New risk factors and uncertainties may emerge from
time to time, and it is not possible for management to predict all
risk factors and uncertainties. Except as required by applicable
law, we do not plan to publicly update or revise any
forward-looking statements contained herein, whether as a result of
any new information, future events, changed circumstances or
otherwise. All forward-looking statements are qualified in their
entirety by this cautionary statement, which is made under the safe
harbor provisions of the Private Securities Litigation Reform Act
of 1995.
This Annual Report on Form 10-K includes our trademarks as well as
trademarks, tradenames and service marks that are the property of
other organizations. Solely for convenience, trademarks and
tradenames referred to in this Annual Report on Form 10-K appear
without the ® and ™ symbols, but those references are not intended
to indicate, in any way, that we will not assert, to the fullest
extent under applicable law, our rights, or that the applicable
owner will not assert its rights, to these trademarks and
tradenames.
We maintain a website at www.erasca.com, to which we regularly post
copies of our press releases as well as additional
information about us. Our filings with the Securities and Exchange
Commission (SEC) are available free of charge through our website
as soon as reasonably practicable after being electronically filed
with or furnished to the SEC. Information contained in our website
does not constitute a part of this report or our other filings with
the SEC.
1
Risk Factor Summary
Below is a summary of the principal factors that make an investment
in our common stock speculative or risky. This summary does not
address all of the risks that we face. Additional discussion of the
risks summarized in this risk factor summary, and other risks that
we face, can be found below under the heading “Risk Factors” in
Item 1A of Part I of this Annual Report on Form 10-K, and should be
carefully considered, together with other information in this
Annual Report on Form 10-K and our other filings with the SEC
before making investment decisions regarding our common
stock.
•
We have a limited operating history, have incurred significant
operating losses since our inception and expect to incur
significant losses for the foreseeable future. We may never
generate any revenue or become profitable or, if we achieve
profitability, we may not be able to sustain it.
•
We will require substantial additional capital to finance our
operations, and a failure to obtain this necessary capital when
needed on acceptable terms, or at all, could force us to delay,
limit, reduce or terminate our development programs,
commercialization efforts or other operations.
•
We are early in our development efforts and are only beginning to
test our product candidates in clinical trials. If we are unable to
successfully develop and commercialize our product candidates or
experience significant delays in doing so, our business will be
materially harmed.
•
Our approach to the discovery and development of product candidates
is unproven, and we do not know whether we will be able to develop
any products of commercial value, or if competing approaches will
limit the commercial value of our product candidates.
•
Clinical and preclinical development involves a lengthy and
expensive process with an uncertain outcome, and the results of
preclinical studies and early clinical trials are not necessarily
predictive of future results. Our product candidates may not have
favorable results in clinical trials, if any, or receive regulatory
approval on a timely basis, or at all.
•
Any difficulties or delays in the commencement or completion, or
termination or suspension, of our current or planned clinical
trials could result in increased costs to us, delay or limit our
ability to generate revenue and adversely affect our commercial
prospects.
•
Use of our product candidates could be associated with side
effects, adverse events or other properties or safety risks, which
could delay or preclude approval, cause us to suspend or
discontinue clinical trials, abandon a product candidate, limit the
commercial profile of an approved label or result in other
significant negative consequences that could severely harm our
business, prospects, operating results and financial
condition.
•
We rely on third parties to conduct many of our preclinical studies
and clinical trials and to manufacture our product candidates, and
these third parties may not perform satisfactorily.
•
We face significant competition, and if our competitors develop
technologies or product candidates more rapidly than we do or their
technologies are more effective, our business and ability to
develop and successfully commercialize products may be adversely
affected.
•
Our business is subject to risks arising from COVID-19 and other
epidemic diseases.
•
Our success depends on our ability to protect our intellectual
property and our proprietary technologies.
•
The trading price of the shares of our common stock could be highly
volatile, and purchasers of our common stock could incur
substantial losses.
2
Item 1. Business.
Overview
At Erasca, our name is our mission: to
erase
cancer.
We are a clinical-stage precision oncology company singularly
focused on discovering, developing, and commercializing therapies
for patients with RAS/MAPK pathway-driven cancers. Molecular
alterations in RAS, the most frequently mutated oncogene, and the
MAPK pathway, one of the most frequently altered signaling pathways
in cancer, account for approximately 5.5 million new patients
diagnosed with cancer globally each year. Our company was
co-founded by leading pioneers in precision oncology and RAS
targeting to create novel therapies and combination regimens
designed to comprehensively shut down the RAS/MAPK pathway for the
treatment of cancer. We have assembled what we believe to be the
deepest, wholly-owned or controlled RAS/MAPK pathway-focused
pipeline in the industry, comprising 11 modality-agnostic programs
aligned with our three therapeutic strategies of: (1) targeting key
upstream and downstream signaling nodes in the RAS/MAPK pathway;
(2) targeting RAS directly; and (3) targeting escape routes that
emerge in response to treatment. The target breadth and molecular
diversity represented in our pipeline enable us to pursue a
systematic, data-driven clinical development effort to identify
single agent and combination approaches with the goal of prolonging
survival in numerous patient populations with high unmet medical
needs.
Our modality-agnostic approach aims to allow us to selectively and
potently inhibit or degrade critical signaling nodes with small
molecule therapeutics, large molecule therapeutics, and protein
degraders. Our purpose-built pipeline includes three clinical-stage
programs (ERK and SHP2 inhibitors, which together comprise our
first, innovative MAPKlamp, and an EGFR inhibitor), one
preclinical-stage program (CNS-penetrant KRAS G12C inhibitor), and
seven discovery-stage programs targeting other key oncogenic
drivers. In 2023, we expect to have four product candidates in the
clinic. In addition, we expect to file an additional
investigational new drug application (IND) every 12-18 months
through 2026. We believe our world-class team’s capabilities and
experience, further guided by our scientific advisory board (SAB),
which includes the world’s leading experts in the RAS/MAPK pathway,
uniquely position us to achieve our bold mission of erasing
cancer.
Of the approximately 5.5 million new patients diagnosed globally
per year with cancers driven by RAS/MAPK pathway molecular
alterations, over 90% have limited or no treatment options. While
the RAS/MAPK pathway has been well characterized and validated
based on multiple compounds approved or in development targeting
discrete signaling nodes in the cascade, most of these compounds
face resistance and tolerability challenges, highlighting the need
for new approaches to target this pathway. We believe that to
effectively shut down a pathway that signals as promiscuously as
RAS/MAPK, a holistic approach must be taken to target not just
individual nodes, but multiple nodes and cooperative mechanisms in
parallel. As depicted in the following figure and described below,
we are pursuing three therapeutic strategies that may be used in
combination with the goal of comprehensively, and perhaps
synergistically, shutting down the RAS/MAPK pathway.
3
1.
Target upstream and downstream MAPK pathway nodes with single
agents and combinations intended to clamp these oncogenic
drivers.
Our first strategy to erase cancer is a novel MAPKlamp that targets
upstream and downstream
nodes, initially SHP2 (ERAS-601) and ERK (ERAS-007), respectively,
to shut down, or “clamp,” the signaling of various oncogenic
drivers, such as receptor tyrosine kinases (RTKs), NF1, RAS, RAF,
and MEK alterations, trapped in between any nodes involving this
pathway. With our MAPKlamp approach, we hope to induce tumor
regression in RAS/MAPK pathway-driven cancers, while also blocking
their main escape routes that lead to tumor resistance. We are also
discovering and developing single agent and combination approaches
to target other upstream nodes that impact the RAS/MAPK pathway
such as EGFR (ERAS-801 and ERAS-12), an RTK that represents a key
escape route for RAS/MAPK signaling, and SOS1 (ERAS-9), a guanine
nucleotide exchange factor that enables RAS to cycle from the
inactive GDP state to the active GTP state.
2.
Target RAS, the midstream MAPK pathway node, directly with single
agents and combinations.
We are discovering and
developing molecules that have the potential to inhibit RAS in its
inactive GDP state (RAS-GDP) as well as its more prevalent active
GTP state (RAS-GTP). Utilizing our in-house discovery efforts
employing structure-based drug design, we are developing
proprietary central nervous system (CNS)-penetrant inhibitors of
KRAS G12C (ERAS-3490), which is the only RAS isoform and mutation
that is more commonly present in the inactive RAS-GDP state. We are
also developing proprietary compounds against KRAS G12D (ERAS-4),
which is more commonly found in the active RAS-GTP state and is the
most prevalent KRAS mutation. Our approach to targeting other RAS
isoforms and mutations that are found more commonly in the RAS-GTP
state is based on the foundational discoveries of one of our
co-founders, Dr. Kevan Shokat, a world-renowned pioneer of novel
therapeutic approaches to targeting key signaling pathways such as
RAS/MAPK in cancer. We entered into an exclusive worldwide license
agreement with University of California San Francisco (UCSF) for
certain intellectual property derived from Dr. Shokat’s work
related to RAS-GTP, which guides our ERAS-2/3
programs.
4
3.
Target escape routes enabled by other proteins or pathways to
further disrupt RAS/MAPK pathway signaling.
RAS-driven
cancers utilize escape routes, namely cooperative mechanisms, to
develop resistance. As an example, RAS-driven cancers can become
dependent on autophagy, which becomes constitutively active and
represents a potential escape route for metabolically active tumors
such as pancreatic ductal adenocarcinoma. By targeting ULK
(ERAS-5), a key regulator of autophagy, in combination with our RAS
targeting agents, we aim to shut down this potential escape route
for RAS-driven cancers. We also are actively pursuing various ways
to further disrupt RAS/MAPK pathway signaling by degrading key
proteins (ERAS-10). Finally, MYC is a transcription factor and
oncogene that is overexpressed in the majority of human cancers and
a key enabler of RAS/MAPK pathway signaling at the transcriptional
level. We are discovering novel approaches to targeting MYC
(ERAS-11).
To pursue these therapeutic strategies, we have assembled and are
developing what we believe is the deepest pipeline targeting
multiple signaling nodes to shut down the RAS/MAPK pathway. We
intend to study these agents either alone or in rational
combinations across multiple relevant tumor types. The following
table summarizes our current, wholly-owned or controlled,
modality-agnostic pipeline to eradicate RAS/MAPK pathway-driven
cancers, and programs that arise from an investment made by Erasca
Ventures, LLC (Erasca Ventures) in a third party.
Our lead product candidates are ERAS-007 (our oral ERK1/2
inhibitor) and ERAS-601 (our oral SHP2 inhibitor), which together
comprise our first MAPKlamp. The extracellular signal-regulated
kinases (ERK), ERK1 and ERK2, belong to a family of
serine-threonine kinases that regulate cellular signaling, and
comprise the terminal node of the RAS/MAPK pathway. ERK proteins
propagate signaling for multiple cellular functions involved in
cell growth and differentiation, which are often overactivated in
RAS/MAPK pathway-driven cancers. We in-licensed ERAS-007 from Asana
BioSciences (Asana) based in part on preclinical studies that
demonstrated the highest potency and longest target residence time
of ERK inhibitors of which we are aware. ERAS-007 has been
evaluated as a single agent in a Phase 1 clinical trial in patients
with advanced solid tumors completed by Asana. Forty-nine patients
were enrolled and administered ERAS-007 once a day (QD) or once
weekly (QW). Objective responses have been observed at doses from
120 mg to 250 mg QW in multiple tumor types (melanoma, salivary
gland tumor, non-small cell lung cancer [NSCLC], and thyroid
cancer) that all harbor alterations (BRAF, HRAS, and NRAS) in the
RAS/MAPK pathway, supporting the development of ERAS-007 QW as a
monotherapy or combination therapy in diverse, biomarker-selected
tumor types. In this trial, ERAS-007 demonstrated a reversible and
manageable adverse event profile.
We are pursuing a broad clinical development plan across multiple
tumor types for ERAS-007, which we refer to as our HERKULES series
of clinical trials, that will include both monotherapy and
combinations with approved and investigational agents, such as RTK,
SHP2, RAS, and/or RAF inhibitors. The first four HERKULES Phase
1b/2 proof-of-concept (POC) clinical trials are exploring both
tissue agnostic and tissue specific indications in patients with
solid tumors and hematologic malignancies, including NSCLC,
colorectal cancer (CRC), and acute myeloid leukemia (AML). In May
2021, we dosed the first patient in HERKULES-1, a Phase 1b/2
clinical trial evaluating ERAS-007 as a single agent and in
combination with ERAS-601 (our first MAPKlamp) in advanced solid
tumors. In September 2021, we dosed the first patient in
HERKULES-2, a Phase 1b/2 master protocol clinical trial for
ERAS-007 and/or ERAS-601 in combination with various agents in
patients with NSCLC. In September 2021, we dosed the first patient
in HERKULES-3, a Phase 1b/2
5
master protocol clinical trial for ERAS-007 in combination with
various agents in patients with GI cancers, with an initial focus
on advanced CRC. In September 2021, we announced a clinical trial
collaboration and supply agreement with Pfizer Inc. (Pfizer) in
connection with the HERKULES-3 trial. Under the terms of the
collaboration, we are sponsoring and funding the clinical trial and
Pfizer is providing its BRAF inhibitor, encorafenib (BRAFTOVI), at
no cost. Additionally, in March 2022, we announced a clinical trial
collaboration and supply agreement with Eli Lilly and Co. (Lilly)
in connection with the HERKULES-3 trial. Under the terms of the
collaboration, we are sponsoring and funding the clinical trial and
Lilly is providing its EGFR antibody, cetuximab (ERBITUX), at no
cost. Finally,
we anticipate dosing
the first patient in HERKULES-4, a Phase 1b/2 master protocol
clinical trial for ERAS-007 and/or ERAS-601 in combination with
various agents in patients with hematologic malignancies. The
master protocols for each of the HERKULES-2, -3 and -4 Phase 1b/2
clinical trials provide the flexibility to explore additional
combinations and expand into other NSCLC, GI cancer and
hematological malignancy indications, respectively. While providing
POC data, these trials may be expanded to enable potential
accelerated approvals in their respective indications.
The second prong of our first MAPKlamp approach, ERAS-601, is
designed to be a potent and selective oral inhibitor of SHP2, a
convergent node for upstream RTK signaling and a critical “on/off
switch” that activates RAS-GTP signaling. SHP2 also drives tumor
cell proliferation and development of resistance. ERAS-601 is
designed to block oncogenic signal transduction and delay the onset
of therapeutic resistance, and thereby serve as a backbone of
combination therapy. In the fourth quarter of 2020, we dosed the
first patient in FLAGSHP-1, a Phase 1 clinical trial for ERAS-601
in patients with advanced solid tumors.
In February 2022, we dosed the first patient in our THUNDERBBOLT-1
Phase 1 clinical trial for ERAS-801, our CNS-penetrant EGFR
inhibitor, in patients with recurrent glioblastoma multiforme
(GBM). In the second half of 2022, we expect to file an IND with
the US Food and Drug Administration (FDA) for ERAS-3490, the
development candidate (DevCan) we nominated from our CNS-penetrant
KRAS G12C inhibitor program. We are also advancing seven other
programs targeting key oncogenic drivers in the RAS/MAPK pathway,
which we will need to successfully progress through discovery and
IND-enabling activities prior to advancing these programs into
clinical development, if at all.
Our core values, team, and social mission
We are a team of experienced drug discoverers, developers, and
company builders who are united by our mission to erase cancer and
passionate about creating potentially life-saving precision
oncology medicines singularly focused on targeting the RAS/MAPK
pathway. Our leadership team has broad and deep experience in
oncology, including advancing therapeutic candidates from discovery
research to clinical development, regulatory approval, and
commercialization. Our core values are embodied by our quest for
the CURE:
Dr. Jonathan Lim, our Chairman, CEO, and Co-Founder, has pioneered
transformative advancements in precision oncology and drug
delivery, including leading Ignyta’s trailblazing pursuit of a
global tissue agnostic label for ROZLYTREK, which became the first
drug in biopharmaceutical history to achieve the unprecedented
triple crown of breakthrough designations with BTD (FDA), PRIME
(EMA) and Sakigake (PMDA). He has served as Chairman and/or CEO and
founding investor of six biotechnology companies that have
collectively achieved global regulatory approval and launch of
seven therapeutic products in oncology, immunology, and drug
delivery, benefitting thousands of patients worldwide.
6
Dr. Michael Varney, our Chair of R&D, SAB member, and a member
of our board of directors, is a pioneer drug discoverer and biotech
leader. His leadership at Agouron resulted in the discovery of
multiple currently marketed anti-cancer agents, including XALKORI
and INLYTA. As Executive Vice President and Head of Genentech’s
Research and Early Development (gRED) and a member of the Roche
Corporate Executive Committee, he was responsible for all aspects
of gRED innovation, drug discovery and development, and built a
team-based organization that today contributes to more than 40% of
Genentech’s development portfolio, including the marketed
anti-cancer agents ERIVEDGE and COTELLIC. Under his leadership,
gRED teams discovered and developed successful medicines that
include VENCLEXTA with AbbVie, the first BCL-2 inhibitor, and
POLIVY, an antibody drug conjugate for the treatment of diffuse
large B-cell lymphoma (DLBCL).
Dr. Wei Lin, our Chief Medical Officer, was responsible for all
development functions and the clinical development of Nektar’s
pipeline, including advancing bempegaldesleukin into multiple
registrational trials and achieving FDA breakthrough therapy
designation in metastatic melanoma. Prior to Nektar, Dr. Lin was
the global development lead in cancer immunotherapy for lung cancer
and head and neck cancer at Roche/Genentech. Under his leadership,
his team oversaw 10 registrational studies, completed five positive
Phase 3 trials, and achieved three US and EU regulatory approvals
for TECENTRIQ, including the first advancement in first-line small
cell lung cancer in three decades. He was also the site head for
oncology product development for Roche China, where his team
achieved multiple additional regulatory approvals for AVASTIN,
ZELBORAF, and TARCEVA.
Dr. David Chacko, our Chief Financial Officer, joined us initially
as Chief Business Officer from Versant Ventures, where he was a
Principal with both investing and operating responsibilities. He
helped lead investment opportunities across multiple therapeutic
areas and advanced several Versant portfolio companies
operationally through company formation, fundraising, corporate and
business development, and clinical and regulatory activities. His
prior roles at Alcon/Novartis, McKinsey, SR One, and Morgan Stanley
bring to Erasca deep experience in strategy, finance, fundraising,
business development, and operations.
Many members of our leadership team have worked together previously
at Ignyta or Roche/Genentech, or have joined us from other leading
companies in the biopharmaceutical and life science tools sectors
such as Aragon, Illumina, Lilly, Medivation, Merck, Myovant,
Neurocrine, Pfizer, Seragon, Synthorx, and Turning Point, and have
worked on numerous oncology drugs that have been approved and
launched for the benefit of patients.
Dr. Lim founded Erasca with Dr. Kevan Shokat (Professor and Chair
of the Department of Cellular and Molecular Pharmacology at UCSF;
Professor of Chemistry at the University of California, Berkeley;
and an investigator at the Howard Hughes Medical Institute), who
sits on our SAB with other RAS/MAPK pathway experts:
•
Dr. Stephen Blacklow is a world expert in SHP2 who helped pioneer
development of the first SHP2 inhibitor with Novartis, and is the
Gustavus Adolphus Pfeiffer Professor of Biological Chemistry and
Molecular Pharmacology, Biological Chemistry and Molecular
Pharmacology at Harvard Medical School; a Professor of Pathology at
the Brigham And Women’s Hospital; a Professor of Cancer Biology at
the Dana-Farber Cancer Institute; and the Chair of the Department
of Biological Chemistry and Molecular Pharmacology at Harvard
Medical School.
•
Dr. Karen Cichowski is a world expert in RAS/MAPK pathway
signaling, including elucidating how deregulated cell signaling
drives tumorigenesis in nervous system, lung, prostate, and breast
cancers, combining translational mouse modeling techniques with
basic biochemical and cell biological studies, and in identifying
novel combination therapies to shut down aberrant RAS/MAPK pathway
signaling. She is Professor of Medicine at Harvard Medical School
and Professor of Medicine/Genetics at Brigham and Women’s
Hospital.
•
Dr. Ryan Corcoran is a gastrointestinal oncologist with a primary
interest in translational oncology research who focuses on targeted
therapies directed against mutations commonly found in human
cancers, such as BRAF and KRAS mutations. He also is a world expert
in ERK, having studied nearly every ERK inhibitor that has been or
is being developed in the field. He is also the Director of the
Gastrointestinal Cancer Center Program; the Scientific Director of
the Termeer Center for Targeted Therapy at Massachusetts General
Hospital Cancer Center; and an Associate Professor of Medicine at
Harvard Medical School.
•
Dr. George Demetri is a world expert in targeted oncology therapies
who pioneered the development of GLEEVEC that helped launch the
revolution in precision oncology. He is the Director of the Center
for Sarcoma and Bone Oncology at the Dana-Farber Cancer Institute;
the Director of the Ludwig Center at the Dana-Farber/Harvard Cancer
Center; and Executive Director for Clinical and Translational
Research at the Ludwig Institute for Cancer Research.
7
•
Dr. Michael Varney is a pioneer drug discoverer and biotech leader
and the former Executive Vice President and Head of Genentech’s
gRED and a former member of the Roche Corporate Executive
Committee.
•
Dr. Pablo Viciana-Rodriguez is a world expert in the RAS/MAPK
pathway whose major focus is the function of the SHOC2 phosphatase
complex as a unique regulatory node required for efficient RAS/MAPK
pathway activation in the context of diseases such as cancer and
RASopathies. He has served as the group leader at the UCL Cancer
Institute since 2008 and is a former postdoctoral researcher in Dr.
Frank McCormick’s lab at the University of California, San
Francisco.
At Erasca, while our mission to erase cancer inspires us, we know
we can do more to make an even broader contribution to society. To
that end, we are pursuing environmental, social, and governance
(ESG) initiatives that are aligned with our core
mission.
•
Erasca Foundation:
In May 2021, we established the Erasca Foundation, which was funded
by the donation of 1,093,557 shares of our common stock (which at
the time represented 1% of our capital stock),
in conjunction with our initial public offering (IPO). The Erasca
Foundation will provide support such as direct research grants,
hardship grants, patient advocacy, patient education in underserved
populations, and funding for other initiatives to positively impact
society.
•
Inclusive clinical trial participation:
We intend to make clinical trials of our product candidates more
accessible to diverse patient
populations and plan to partner with others who are like-minded in
this regard.
•
Drug access program:
We intend to provide patients with access to the drugs we develop
and commercialize, including through
compassionate use programs if our products are demonstrated to be
safe and efficacious. We also intend to increase access to
life-changing drugs in underserved populations if our products
become commercially available.
Our corporate strategies to erase cancer
Our mission is to erase cancer by eradicating RAS/MAPK
pathway-driven cancers. Our corporate strategies to achieve our
mission include:
•
Relentlessly focus on patients and society in our mission to erase
cancer.
There are approximately 5.5 million new patients
diagnosed globally per year with cancers driven by RAS/MAPK pathway
alterations, over 90% of whom have limited or no treatment options.
We are a team of experienced drug discoverers, developers, and
company builders who are united by our mission to erase cancer and
passionate about creating potentially life-saving precision
oncology medicines. In addition, we are pursuing ESG initiatives
that are aligned with our core mission.
•
Develop novel single agent and combination regimens to
comprehensively shut down the RAS/MAPK pathway for the treatment of
cancer.
We are pursuing three therapeutic strategies that may be used in
combination to comprehensively, and perhaps
synergistically, shut down the RAS/MAPK pathway: (1) target
upstream and downstream MAPK pathway nodes with single agents and
combinations intended to clamp these oncogenic drivers; (2) target
RAS directly with single agents and combinations; and (3) target
escape routes enabled by other proteins or pathways to further
disrupt RAS/MAPK pathway signaling. Our strategic focus on the
RAS/MAPK pathway allows us to comprehensively target every critical
node in the pathway that could drive cancer signaling.
•
Advance our deep, modality-agnostic RAS/MAPK pathway-focused
pipeline.
We believe our internally and externally sourced
RAS/MAPK pathway-focused pipeline, comprising 11 targeted therapy
programs, is the deepest in the industry. Our modality-agnostic
approach aims to selectively and potently inhibit or degrade
critical RAS/MAPK signaling nodes with small molecule therapeutics,
large molecule therapeutics, and protein degraders. ERAS-007 (our
ERK inhibitor), ERAS-601 (our SHP2 inhibitor), and ERAS-801 (our
EGFR inhibitor) are currently being studied in clinical trials. In
2023, we expect to have four product candidates in the clinic. In
addition, we expect to file an additional IND every 12-18 months
through 2026. Given the high unmet medical need of the patients we
seek to treat, we will evaluate the potential for expedited
development and review pathways.
8
•
Internally and externally source, on a global basis, potentially
disruptive programs targeting RAS/MAPK pathway alterations.
We have built a productive and efficient internal discovery engine.
Our world-class structural biology team generates more
than 125 protein structures annually and we use computational
biology and computational chemistry to accelerate our discovery
activities. While we have strong internal capabilities, we also
believe that innovation is a collective, global endeavor and a
single platform is unlikely to discover all the best ideas and
approaches. We therefore plan to continue to pursue synergistic,
in-pathway opportunities, regardless of origin, that meet our high
scientific bar. Our extensive network and relationships provide us
preferential—and at times exclusive—access to certain assets of
interest.
•
Lead the next revolution in precision oncology.
The first wave of precision oncology included tyrosine kinase
inhibitors such as
ROZLYTREK, approved for select tumors that harbor ROS1 or NTRK
fusions. While these initial development efforts focused on
specific disease-causing alterations in areas of high unmet medical
need, these patient populations were modest in size. We believe
that to effectively shut down a pathway that signals as
promiscuously as RAS/MAPK and that encompasses a range of
alterations, a holistic approach must be taken to target not just
specific individual mutations, but multiple alterations and
cooperative mechanisms in parallel. We are pursuing tissue agnostic
and tissue specific labels with dynamically designed
biomarker-based basket and umbrella studies, respectively, as well
as master protocols, in order to quickly demonstrate clinical
proof-of-concept in a variety of tumor types for both single agent
and combination approaches.
•
Evaluate opportunities to accelerate development timelines and
enhance the commercial potential of our programs in collaboration
with third parties.
We own or control worldwide development and commercialization
rights to our entire pipeline of 11
targeted therapy programs (excluding programs in our pipeline that
arise from an investment made by Erasca Ventures in a third party).
This provides us with the flexibility to explore combinations of
our agents with each other, other investigational agents, and/or
standard of care therapies. We intend to continue evaluating
opportunities to work with partners that meaningfully enhance our
capabilities with respect to the development and commercialization
of our product candidates. In addition, we intend to commercialize
our product candidates in the United States and possibly Europe,
where as many as 1.8 million patients are diagnosed annually with
RAS/MAPK pathway alterations. We intend to explore partnerships in
selected geographies to maximize the worldwide commercial potential
of our programs.
Our singular focus on the RAS/MAPK pathway
Background
The RAS/MAPK pathway is one of the most frequently altered
signaling pathways in cancer. Molecular alterations in key
signaling nodes within the RAS/MAPK pathway have been shown to
drive cell proliferation across a wide range of tumor types. As
described further below, our wholly-owned or controlled pipeline
targets all of the key signaling nodes colored in purple, either
directly or indirectly as single agents and in combination in order
to prolong survival in a wide range of patient
populations.
9
EGFR/FLT3
EGFR and FLT3 are RTKs, which are proteins that are embedded in the
cell membrane and relay growth signals from the outside environment
to the cell’s internal machinery. At rest, these proteins reside on
the cell membrane as inactive monomers. Growth factors secreted by
nearby cells bind to specific RTKs, such as growth factor EGF
binding to EGFR and FL binding to FLT3, and cause these RTKs to
dimerize. Dimerized RTKs activate one another through
transphosphorylation of their intracellular regions. Intracellular
proteins, such as adapter proteins, bind to these phosphorylated
regions and propagate the pro-growth signals within the cell via
one or more signaling pathways. Cells express a variety of RTKs so
that environmental cues can be relayed to specific cell populations
in specific contexts. EGFR mediates pro-growth signaling in skin
and in the ducts and outer surfaces of many organs. FLT3
predominantly mediates pro-growth signaling in immature blood
cells.
Overactive RTK signaling can result in uncontrolled cell growth and
survival that transforms normal cells into cancer cells.
SHP2
SHP2 is a protein tyrosine phosphatase and a key positive regulator
of the growth signals from the RTK growth factor receptors to the
intracellular signaling pathways (including RAS/MAPK and PI3K) that
promote growth and survival of normal cells and cancer cells. As
such, SHP2 is a convergent node for upstream RTK signaling:
activated SHP2 upregulates (“turns up”) the positive signals and
downregulates (“turns down”) the negative signals in the signaling
cascades. SHP2 also serves as a central node in relaying the growth
and survival signals from RTKs such as EGFR and FLT3 to RAS/MAPK
and other intracellular pathways. SHP2 is an attractive target
because SHP2 inhibition ubiquitously blocks the growth signals from
multiple RTKs, preventing cancer cells from bypassing the blockade
on a specific RTK (e.g., EGFR inhibitor) through activation of
other RTK growth factor receptors (e.g., MET).
10
SOS1
SOS1, or Son of Sevenless-1, is a protein that is recruited to RTK
complexes and, in turn, recruits and activates members of the RAS
protein family. SOS1 activates RAS proteins by acting as a guanine
nucleotide exchange factor (GEF), which facilitates the exchange of
the RAS-bound nucleotide from guanine diphosphate (GDP) to guanine
triphosphate (GTP). When GDP is exchanged for GTP, RAS adopts an
active conformation that enables it to bind and activate downstream
effector proteins, such as the RAF family, which ultimately results
in RAS/MAPK pathway activation.
NF1
NF1, or neurofibromin, is a protein that accelerates the transition
of RAS proteins from the active RAS-GTP state to the inactive
RAS-GDP state. NF1 is classified as a GTPase activating protein
(GAP) because it boosts the ability of RAS to hydrolyze bound GTP
to GDP. Although RAS can autonomously hydrolyze GTP, it is
dependent on GAPs such as NF1 to rapidly cycle it from the active
state to the inactive state and thereby prevent overactive
signaling. If NF1 is inactivated due to a mutation (NF1
loss-of-function mutation), RAS proteins may spend more time in the
active RAS-GTP state. This can result in hyperactive RAS/MAPK
pathway activation that drives aberrant cell growth and ultimately
tumorigenesis. This is observed in patients affected by a genetic
disorder caused by somatic mutations in the NF1 gene called
neurofibromatosis type 1. NF1 loss-of-function mutations are
observed in a variety of cancers, including melanoma and CRC, where
they activate RAS/MAPK signaling alone or in conjunction with other
RAS/MAPK pathway activating mutations.
RAS
RAS proteins are ubiquitously expressed GTPase proteins. The RAS
protein family consists of KRAS, NRAS, and HRAS proteins and acts
as the entry node in the MAPK signaling pathway. KRAS is the most
abundantly expressed RAS protein followed by NRAS and then HRAS.
RAS proteins act as signaling transducers since they are recruited
to activated RTK complexes where they are converted into an active
conformation (RAS-GTP) that enables them to activate downstream
effector proteins, such as RAF proteins. The activation state of a
RAS protein is dictated by the phosphorylation state of the bound
guanosine: RAS adopts an inactive RAS-GDP conformation when bound
to GDP and an active RAS-GTP conformation when bound to GTP.
Conversion of RAS into an active conformation is mediated by
binding to co-factor proteins, e.g., SOS1, and these co-factor
proteins enable the exchange of the RAS-bound nucleotide from GDP
to GTP. In the active state, RAS-GTP proteins interact with
multiple effector proteins to propagate cell signaling through
multiple pathways. For example, activated RAS-GTP proteins interact
with RAF proteins to activate MAPK signaling, and PI3K proteins to
activate PI3K pathway signaling. RAS can transition from the active
state into the inactive state by hydrolyzing its bound nucleotide
from GTP to GDP either intrinsically or catalyzed through
interactions with co-factor proteins, such as NF1. RAS proteins are
the most frequently mutated oncoproteins in cancer. These mutations
occur at hotspots, such as amino acid residues 12, 13, and 61, and
these hotspot mutations impair RAS’s ability to hydrolyze GTP to
GDP. As a result, mutant RAS-GTP remains in the active state for
prolonged periods of time resulting in hyperactive stimulation of
the RAS/MAPK and other pathways.
RAF
RAF proteins are ubiquitously expressed serine-threonine kinases
that are a part of the RAS/MAPK pathway and whose activity is
regulated by RAS proteins. The RAF protein family consists of ARAF,
BRAF, and CRAF (RAF1). In the absence of activated RAS-GTP, RAF
proteins assume an autoinhibited conformation in complex with
downstream effector proteins, MEK1 and MEK2. RAF proteins can
homodimerize (e.g., BRAF-BRAF dimers) or heterodimerize (e.g.,
CRAF-BRAF dimers). When RAF proteins bind to activated RAS-GTP,
they adopt an active conformation that results in activation of
their kinase domains. The activated kinase domains then
phosphorylate complexed MEK proteins, activating those proteins and
releasing them from the RAF-MEK complex. Activated MEK then signals
further down the RAS/MAPK pathway. Mutations in RAF proteins,
especially in BRAF, have been observed in many cancers, such as
melanoma, CRC, NSCLC, and thyroid cancer. For example, the BRAF
V600E mutation (a class I BRAF mutation) is frequently observed in
melanoma and this mutation enables BRAF to constitutively activate
MEK as a monomer. Approved BRAF inhibitors for class I mutations
include vemurafenib, dabrafenib, and encorafenib. Class II BRAF
mutations enable BRAF to constitutively dimerize and activate MEK.
Class III BRAF mutations impair the ability of the mutant BRAF
protein to phosphorylate MEK, but class III mutant BRAF proteins
can aberrantly dimerize with wildtype RAF proteins and enable their
dimerized wildtype RAF partners to activate MEK. To our knowledge,
there are no approved inhibitors of BRAF Class II or Class III
mutations. A number of inhibitors targeting BRAF Class II and Class
III mutations, as well as pan-RAF inhibitors designed to disrupt
wildtype RAF signaling, are in development; however, to our
knowledge, none have received regulatory approval.
11
MEK
MEK1 and MEK2 proteins are ubiquitously expressed serine-threonine
kinases that are activated by RAF-mediated phosphorylation and
signal downstream by activating ERK proteins. MEK1 and MEK2
proteins form complexes with RAF proteins in the inactive state and
are recruited as a unit to activated RAS-GTP. RAS-GTP then
activates the RAF-MEK complex by binding to RAF, which then
activates MEK via phosphorylation and releases from the RAF-MEK
complex. Activated MEK then selectively phosphorylates ERK1 and
ERK2 proteins, which are the terminal nodes of the RAS/MAPK
pathway. Currently approved MEK inhibitors, such as trametinib,
binimetinib, cobimetinib, and selumetinib, allosterically bind MEK
proteins and inhibit MEK activation, either as free proteins alone
or in complex with RAF. The inhibition of RAS/MAPK signaling by MEK
inhibitors can result in an upregulation of signaling upstream of
MEK due to negative feedback loops within the RAS/MAPK pathway.
This increased signaling pressure can overwhelm MEK inhibitors and
result in reactivation of MAPK signaling. Most MEK inhibitors are
approved in combination with a BRAF inhibitor partially due to
their vulnerability of being overwhelmed by the reactivation of
MAPK signaling. In this combination, BRAF inhibitors attenuate
upstream signaling pressure on MEK inhibitors, and MEK inhibitors
further limit downstream MAPK signaling not inhibited by the BRAF
inhibitor.
ERK
The extracellular signal-regulated kinases (ERK), ERK1 and ERK2,
are ubiquitous serine-threonine kinases that regulate cellular
signaling in both physiological and pathological states and
comprise the terminal node of the RAS/MAPK pathway. Once activated
by MEK, ERK proteins phosphorylate thousands of downstream
proteins, propagating RAS/MAPK signaling across multiple cellular
functions. In contrast to currently approved allosteric MEK
inhibitors, ERK inhibitors in development are ATP-competitive and
as a result, their potency is robust against the activated state of
ERK. Based on this property, ERK inhibitors potentially can
overcome drug resistance mechanisms that involve reactivation of
RAS/MAPK pathway signaling, such as a rebound of RAS/MAPK signaling
resulting from the alleviation of negative feedback or an upstream
RAS/MAPK pathway protein adopting an acquired resistance
mutation.
ULK
Autophagy is a metabolic process that cells use to break down and
recycle cellular components. This process enables cells to renew
cellular components whose functions are impaired due to age or
malfunction. Autophagy also serves as a survival mechanism in
nutrient deprived conditions, enabling the cell to continue to
synthesize critical cellular components. RAS-driven tumor cells
reconfigure many of their metabolic processes, including autophagy,
to better fuel their growth and survival. Autophagy can act as a
resistance mechanism to RAS/MAPK pathway inhibitors by becoming
constitutively active and enabling the cell to survive metabolic
stresses induced by the inhibition of RAS/MAPK pathway signaling.
ULK1 and ULK2 are serine-threonine kinases that control the
initiation of autophagy, thereby acting as gatekeepers of
autophagy. Combining a RAS/MAPK pathway inhibitor with an ULK1
and/or ULK2 inhibitor can potentially inhibit tumor growth by
blocking upregulation of autophagy, a potential escape route or
adaptive resistance mechanism to the RAS/MAPK pathway
inhibitor.
MYC
The MYC protein (also known as c-MYC) is a transcription factor
that regulates the transcription of hundreds of genes that are
associated with cell growth. Transcription factors guide the
cellular machinery to transcribe specific genes in the nucleus and
those transcribed genes are then translated into proteins in the
cytoplasm. Transcription factors are regulated by multiple
signaling pathways, including RAS/MAPK, and they integrate this
signaling information to transcribe genes in a context-dependent
manner. Dimerization is required for MYC transcription factor
activity and MYC’s most frequent dimerization partner is MAX. The
activity of the MYC-MAX complex is largely driven by the
concentration of MYC protein in the nucleus but other factors, such
as the phosphorylation status of MYC, also regulate MYC activity.
Given MYC’s critical role in regulating genes that drive cell
growth, MYC function is dysregulated in 40% of cancers and MYC
overexpression is the most frequent form of MYC dysregulation.
MYC’s role as a transcription factor and not an enzyme has made the
development of inhibitors targeting the MYC protein challenging. We
believe inhibiting the transcription of MYC and/or MYC-agonists,
such as MAX, offers a promising alternative therapeutic approach to
reduce MYC activity in tumors where the traditional direct
targeting of the MYC protein has failed. MYC and RAS are two of the
most commonly dysregulated genes in human cancer and are also
downstream effectors for a range of other oncogenic mutations in a
variety of tumor types. MYC and RAS also frequently cooperate with
each other in tumor development, heightening the urgency of
targeting these two pivotal oncogenes.
12
Our approach is focused on comprehensively silencing the RAS/MAPK
pathway by targeting these key signaling nodes, from upstream RTKs
to downstream nuclear transcription factors, which have been shown
to drive cell proliferation across a wide range of tumor
types.
Patient lives at stake annually with RAS/MAPK pathway
alterations
At Erasca, we are on a bold mission to erase cancer. The journey
will be long, and it won’t be easy. But patients with cancer are
waiting, and we are eager to make new therapies available as soon
as possible. Our mission will involve delivering new therapies to
patients in markets where there are limited or no approved
therapies, which are referred to as “blue oceans” (adapted
from
Blue Ocean Strategy
by Chan Kim & Renée Mauborgne), as well as markets where there
are already approved or soon to be approved product offerings, or
“red oceans.” Of the approximately 5.5 million new patients
diagnosed globally per year with cancers driven by RAS/MAPK pathway
alterations, over 90% (approximately 5 million patients) are in
blue oceans with limited or no treatment options. In the United
States and Europe, there are over 1.8 million patients per annum
who could be treated with the therapies we are seeking to develop
and commercialize. In other parts of the world, we intend to
explore partnerships in selected geographies to maximize the
worldwide commercial potential of our programs.
Our therapeutic strategies for shutting down the RAS/MAPK
pathway
We believe that to effectively shut down a pathway that signals as
promiscuously as RAS/MAPK, a holistic approach must be taken to
target not just single nodes, but multiple nodes and cooperative
mechanisms in parallel. We believe our internally and externally
sourced RAS/MAPK pathway-focused pipeline, comprising 11 targeted
therapy programs, is the deepest in the industry. The target
breadth and molecular diversity represented in our pipeline enable
us to pursue a systematic, data-driven clinical development effort
to identify single agent and combination approaches that aim to
prolong survival in numerous patient populations with high unmet
medical needs. We are pursuing three therapeutic strategies that
may be used in combination with the goal of comprehensively, and
perhaps synergistically, shutting down the RAS/MAPK
pathway:
13
1.
Target upstream and downstream MAPK pathway nodes with single
agents and combinations intended to clamp these oncogenic
drivers.
Our first therapeutic strategy to erase cancer is a novel MAPKlamp
that targets upstream and
downstream nodes, initially SHP2 and ERK, respectively, to shut
down, or clamp, the signaling of various oncogenic drivers such as
RTKs, NF1, RAS, RAF, and MEK alterations trapped in between any
nodes involving this pathway. With our MAPKlamp approach, we aim to
induce tumor regression in RAS/MAPK pathway-driven cancers, while
also blocking their main escape routes that lead to tumor
resistance. We are also discovering and developing single agent and
combination approaches to target other upstream nodes that impact
the RAS/MAPK pathway such as EGFR, an RTK that represents a key
escape route for RAS/MAPK pathway signaling, and SOS1, a GEF that
enables RAS to cycle from the inactive GDP state to the active GTP
state.
2.
Target RAS, the midstream MAPK pathway node, directly with single
agents and combinations.
Our second therapeutic
strategy to erase cancer is to target RAS directly by discovering
and developing molecules that have the potential to inhibit
RAS-GDP, as well as the more prevalent RAS-GTP. Utilizing our
in-house discovery efforts employing structure-based drug/degrader
design (SBDD), we are developing CNS-penetrant inhibitors of KRAS
G12C, which is the only RAS isoform and mutation that is more
commonly present in the inactive RAS-GDP state. We are also
employing SBDD to develop proprietary compounds against KRAS G12D,
which is more commonly found in the active RAS-GTP state and is the
most prevalent KRAS mutation. Our approach to targeting other RAS
isoforms and mutations that are found more commonly in the RAS-GTP
state is based on the foundational discoveries of one of our
co-founders, Dr. Kevan Shokat, a world-renowned pioneer of novel
therapeutic approaches to targeting key signaling pathways such as
RAS/MAPK in cancer.
3.
Target escape routes enabled by other proteins or pathways to
further disrupt RAS/MAPK pathway signaling.
Our third
therapeutic strategy to erase cancer is to target other pathways
and mechanisms that cooperate with RAS/MAPK pathway signaling. For
example, RAS-driven cancers can become dependent on autophagy,
which becomes constitutively active and represents a potential
escape route for metabolically active tumors such as pancreatic
ductal adenocarcinoma. By targeting ULK, a key regulator of
autophagy, in combination with our RAS targeting agents, we aim to
shut down this potential escape route for RAS-driven cancers. We
also are actively pursuing various ways to further disrupt RAS/MAPK
signaling by degrading key proteins. Finally, MYC is a
transcription factor and oncogene that is overexpressed in the
majority of human cancers, contributing to at least 40% of tumors
and is a key enabler of RAS/MAPK pathway signaling at the
transcriptional level. We are discovering novel approaches to
targeting MYC.
Our strategic focus on the RAS/MAPK pathway allows us to
comprehensively target every critical node in the pathway that
could drive signaling. As shown in the figure below, our
wholly-owned or controlled pipeline targets, either directly or
indirectly, each of the signaling nodes colored in
purple.
14
Our innovation model
Due to the magnitude of the challenge of erasing cancer, we are
combining our robust internal discovery and development
capabilities with a global in-licensing and acquisition strategy to
assemble the industry’s deepest, modality-agnostic RAS/MAPK
pathway-focused pipeline. We believe these complementary approaches
to innovation provide us with important optionality, both
therapeutically and strategically, as we endeavor to bring forth
the next generation of potentially differentiated targeted
therapies for RAS/MAPK pathway-driven cancers.
Internal discovery and development
We have built a productive and efficient internal discovery engine
at the heart of which lies SBDD, a key tool for the discovery of
novel small molecule therapeutics and protein degraders by
elucidating the three-dimensional structure of the potential drug
molecule or degrader bound to the target protein of interest,
allowing scientists to better understand and iterate on the
structure-activity relationship of their hit and lead compounds or
degraders. Three of our senior scientists were early pioneers in
the use of SBDD while at Agouron Pharmaceuticals (now Pfizer), the
first biotechnology company to use protein structure to inform
medicinal chemistry for drug discovery: Dr. Dave Matthews, the
scientific founder of Agouron, is our Senior Crystallography
Advisor; Dr. Michael Varney, one of the original employees at
Agouron who built the team that developed SBDD, is our Chair of
R&D, SAB member, and a member of our board of directors; and
Dr. Ping Chen, our Senior Director of Crystallography, was a member
of Dr. Matthews’ structural biology team at Agouron, and joined us
from Pfizer to lead our internal structural biology efforts which
generates more than 100 protein structures annually to guide our
discovery research.
15
We use computational biology and computational chemistry to
accelerate our discovery activities. We have standardized how we
characterize our compounds across in vitro/vivo activity, drug
distribution, metabolism, and pharmacokinetics (PK), structural,
and secondary pharmacology assays, and centralized the storage of
these data for automated analyses. These data are continuously
reviewed by our scientific teams, and promising trends, including
unpredicted ones that arise serendipitously, are prioritized for
future exploration.
We supplement our medicinal chemistry efforts with fragment
screens, including DNA encoded library (DEL) screens, to identify
novel chemical matter with promising activity against targets of
interest. These “hits” give us starting points for our early-stage
drug discovery programs, and also provide opportunities to
diversify molecular designs for later-stage discovery programs. DEL
screens interrogate the binding of billions of compounds against
our targets and increase the likelihood that we will discover a
fragment that we can eventually transform into a potent
therapy.
Based on our previous collective experiences at Ignyta,
Roche/Genentech, Pfizer, and elsewhere, our team has extensive
precision oncology expertise with dynamic clinical trial designs
such as adaptive trials, biomarker-based basket and umbrella
studies, and master protocols. We will continue to leverage this
experience, in collaboration with industry and academic partners,
in order to quickly demonstrate clinical proof-of-concept in a
variety of tumor types for both single agent and combination
approaches.
External sources of innovation
We believe innovation in cancer therapy is a collective, global
endeavor unlikely to emerge from a single company or a single
platform. There are exciting product candidates, technologies, and
approaches in development worldwide, and our innovation model gives
us the flexibility to supplement our internal efforts with
externally sourced assets through collaboration, in-license, or
acquisition. We also established Erasca Ventures, our wholly-owned
subsidiary, in March 2021 to make equity investments in early-stage
biotechnology companies that are aligned with our mission and
strategy. In March 2022, Erasca Ventures made an equity investment
in Affini-T Therapeutics, Inc. (Affini-T) to develop Affini-T’s
potential best-in-class T-cell receptor (TCR) cell therapies
targeting multiple oncogenic driver mutations, including KRAS G12V
and KRAS G12D. To date, we have in-licensed or acquired novel
therapies from multiple geographic regions, including our
clinical-stage, oral ERK1/2 inhibitor, ERAS-007, which we acquired
from Asana.
We leverage our extensive network of preferred relationships with
our Scientific and Research & Development Advisory Boards, as
well as leading institutional investors, investment banks, academic
institutions, and biopharmaceutical companies that keep us apprised
of assets of strategic interest. We pursue the best science in the
world, regardless of its origin, and will continue to evaluate
additional opportunities to strengthen and diversify our pipeline
through academic and biopharmaceutical collaborations, in-licenses,
acquisitions, and strategic investments that meet our high
scientific bar and can help us advance our mission to erase
cancer.
Modality-agnostic pipeline
Cancer is a complex, heterogeneous disease that is unlikely to
succumb to a one-size-fits-all approach. We believe shutting down
the RAS/MAPK pathway in cancer requires a systematic, data-driven
approach to development, part of which involves choosing the most
appropriate technology for the target of interest, or what we call
a modality-agnostic approach. We therefore seek to understand the
biology of the target of interest first, and then choose the
therapeutic modality best suited to optimally inhibit or degrade
that target. We are currently utilizing several modalities to
target the RAS/MAPK pathway, including small molecule therapeutics,
large molecule therapeutics, and protein degraders.
For example, we are developing protein degraders in addition to our
internal small molecule discovery capabilities, including
proteolysis targeting chimeras (PROTACs), as a complementary
strategy to modulate RAS/MAPK pathway proteins of interest.
PROTAC-mediated degradation is a viable option for attenuating
oncogenic RAS/MAPK pathway levels and downstream signaling in
cancer cells, by utilizing the body’s own natural disposal system
to remove oncogenic proteins selectively and
efficiently.
16
Our pipeline
We have assembled what we believe is the deepest, wholly-owned or
controlled RAS/MAPK pathway-focused pipeline in the industry,
comprising 11 modality-agnostic programs aligned with our three
therapeutic strategies of: (1) targeting key upstream and
downstream signaling nodes in the RAS/MAPK pathway; (2) targeting
RAS directly; and (3) targeting escape routes that emerge in
response to treatment. The table below summarizes our current
pipeline. We have exclusive worldwide development and commercial
rights for all of our programs (excluding programs in our pipeline
that arise from an investment made by Erasca Ventures in a third
party).
Importantly, we believe the target breadth and molecular diversity
represented in our pipeline enable us to pursue a systematic,
data-driven clinical development effort to identify single agent
and combination approaches designed to comprehensively shut down
the RAS/MAPK pathway in a range of underserved cancer indications.
The figure below illustrates the overlay between our current
pipeline and several key nodes in the RAS/MAPK pathway that we
believe are attractive targets for therapeutic intervention. Our
pipeline also provides potential solutions for patients with
limited or no treatments available by directly targeting with
single agents and/or clamping with combinations, the various nodes
of the RAS/MAPK pathway.
17
MAPKlamp: our therapeutic strategy targeting proximal and terminal
nodes of the RAS/MAPK pathway
Our first therapeutic strategy to erase cancer with various
combinations is MAPKlamp, our novel approach targeting upstream and
downstream nodes in the RAS/MAPK pathway designed to shut down, or
clamp, the signaling of various oncogenic drivers, such as RTKs,
NF1, RAS, RAF, and MEK alterations trapped in between any nodes
involving this pathway. With our MAPKlamp approach, we aim to
induce tumor regression in RAS/MAPK pathway-driven cancers, while
also blocking the main escape routes that lead to tumor
resistance.
Our lead product candidates are ERAS-007 (our oral ERK1/2
inhibitor) and ERAS-601 (our oral SHP2 inhibitor), which together
comprise our first, innovative MAPKlamp approach. ERK and SHP2 are
the convergent downstream and upstream nodes of the RAS/MAPK
pathway, respectively. ERK proteins propagate signaling for
multiple cellular functions involved in cell growth and
differentiation, which are often overactivated in RAS/MAPK
pathway-driven cancers. We believe that targeting ERK, the terminal
node of the RAS/MAPK pathway, is preferable to targeting MEK
because it is less prone to MAPK pathway reactivation, which leads
to greater suppression of signaling. The second prong of our first
MAPKlamp, ERAS-601, is a potent and selective oral inhibitor of
SHP2, a critical “on/off switch” that activates RAS-GTP signaling.
Our SHP2 inhibitor is designed to block oncogenic signal
transduction and delay the onset of therapeutic resistance.
Targeting either or both of these key nodes thereby has the
potential to serve as a backbone of combination therapy against
RAS/MAPK pathway altered cancers.
As our portfolio advances, we anticipate additional MAPKlamp
combinations to emerge. Given the breadth of our pipeline, we
believe we are the only company that has the therapeutic and
strategic flexibility to comprehensively target every critical node
in the RAS/MAPK pathway that could drive cancer
signaling.
ERAS-007: our ERK inhibitor
ERAS-007 is designed to be a potent and selective oral inhibitor of
ERK1/2. We in-licensed ERAS-007 from Asana based in part on
preclinical studies that demonstrated the highest potency and
longest target residence time of ERK inhibitors of which we are
aware. In a Phase 1 clinical trial completed by Asana, ERAS-007
demonstrated single-agent activity including objective responses in
tumors harboring RAS/MAPK pathway alterations and was well
tolerated. We are pursuing a broad clinical development plan across
multiple tumor types for ERAS-007, which we refer to as our
HERKULES series of clinical trials, that will include both
monotherapy and combinations with approved and investigational
agents, such as RTK, SHP2, RAS, and/or RAF inhibitors. The first
four HERKULES Phase 1b/2 clinical trials are exploring both tissue
agnostic and tissue specific indications in patients with solid
tumors and hematologic malignancies. We dosed the first patient in
HERKULES-1 in May 2021 and the first patients in HERKULES-2 and
HERKULES-3 in September 2021. We believe that as many as 4.5
million patients worldwide per year could benefit from ERAS-007
combinations that include MAPKlamp, including 4.0 million patients
with blue ocean indications where there are currently limited or no
approved therapies.
Preclinical profile of ERAS-007
Asana completed a series of preclinical studies to characterize the
differentiated attributes, namely high potency and long target
residence time, of ERAS-007 in vivo and in vitro. In multiple
assays, ERAS-007 achieved potent, reversible, and ATP-competitive
inhibition of ERK1 and ERK2 with a biochemical IC50 (a measure of
50% inhibition) against both ERK1 and ERK2 of 2 nM and cell-based
mechanistic IC50 against pRSK of 7 nM. In addition, ERAS-007
exhibited long biochemical residence time while bound to ERK, which
has been measured as 550 minutes against ERK2. This longer target
residence time compared to other clinical-stage ERK inhibitors may
allow for longer intervals between doses in patients.
18
ERAS-007 IC50s against ERK1 and ERK2 were characterized in a
biochemical kinase activity. Cell-based IC50 was characterized by
the ability of ERAS-007 to inhibit ERK from phosphorylating one of
its downstream targets, RSK1. pRSK represents RSK1
phosphorylation.
The biochemical binding properties of three clinical-stage ERK
inhibitors shown as both the rate at which each inhibitor
dissociates from ERK2 (koff)
and the period of time that each ERK inhibitor binds ERK2
(residence time).
This biochemical potency has translated into strong
anti-proliferative activity in cell lines with mutations in the
RAS/MAPK pathway compared to other clinical-stage ERK inhibitor
compounds. In 14 out of 14 cell lines that harbored activating
RAS/MAPK pathway alterations, ERAS-007 exhibited potent activity
with a less than 1 µM IC50. In two KRAS G12C cell lines, ERAS-007
showed greater potency compared to ulixertinib, an ERK inhibitor,
comparable potency to binimetinib, a MEK inhibitor, and sotorasib,
a KRAS G12C inhibitor. Cellular signaling studies demonstrated that
ERAS-007 inhibited phosphorylation of downstream targets of ERK
such as ribosomal S6 kinases (RSK), Fos-related antigen (FRA), and
ETS domain-containing protein (ELK) in the BRAF V600E CRC HT-29
cell line. Demonstrating its selectivity, in seven out of eight
cell lines that did not harbor any activating RAS/MAPK pathway
alterations, ERAS-007 showed weak inhibition with a greater than 10
µM IC50. Together, these results suggest that ERAS-007 is a potent
and selective ERK inhibitor with the ability to inhibit cell growth
in multiple models of RAS/MAPK pathway-driven cancers relative to
other agents used in these settings.
ERAS-007, ulixertinib and ravoxertinib were profiled in 3 BRAF
mutant, 1 HRAS mutant, 8 KRAS mutant, 2 NRAS mutant, and 8 wildtype
RAS and wildtype RAF cell lines. Nanomolar IC50 values are
represented on the y-axis. Lower IC50s denote stronger
activity.
ERAS-007 inhibits cell viability of two KRAS G12C mutant cell
lines, pancreatic carcinoma MIA PaCa-2 and NSCLC NCI-H358, with
higher potency than a clinical-stage ERK inhibitor, ulixertinib,
and comparable potency to an approved MEK inhibitor, binimetinib,
and an approved KRAS G12C inhibitor, sotorasib.
19
Inhibition of signaling by kinases is typically achieved by either:
(1) ATP-competitive inhibition whereby an inhibitor blocks ATP
binding or (2) allosteric inhibition whereby an inhibitor does not
block ATP binding but rather binds to a different region to prevent
the kinase from signaling downstream. Currently approved MEK
inhibitors, trametinib, binimetinib, selumetinib, and cobimetinib,
are allosteric MEK inhibitors. A potential limitation of these
allosteric MEK inhibitors is that they preferentially bind MEK in
the inactive state and have weaker inhibitory activity against
activated MEK proteins. Another limitation is that some MEK
inhibitors preferentially disrupt activation via one RAF family
member (e.g., BRAF) but not another (e.g., CRAF). Due to negative
feedback regulation in the RAS/MAPK pathway, inhibition of
downstream signaling nodes can result in RAS/MAPK pathway feedback
reactivation that is mediated through multiple members of the RAF
family. This increased upstream signaling pressure can serve as a
resistance mechanism to MEK inhibitors and has been observed in the
clinic. As an ATP-competitive ERK inhibitor, ERAS-007 has been
shown to more robustly block RAS/MAPK pathway reactivation than
allosteric MEK inhibitors. As shown in the figure below, ERAS-007
continuously inhibited downstream ERK activity in a KRAS G12C
mutant CRC cell line, whereas the RAS/MAPK pathway was reactivated
beginning as early as 24 hours after treatment with each of the
three MEK inhibitors, which is illustrated with the emergence of
the dark P-RSK bands (darker intensity equates to higher signaling
or reactivation) in the following Western blots.
Western blot characterization of three MEK inhibitors (selumetinib,
binimetinib, and trametinib) and two ERK inhibitors (ulixertinib
and ERAS-007) in the KRAS G12C mutant CRC cell line SW1463. The
phosphorylation states of RSK (P-RSK), ERK (P-ERK), CRAF (P-CRAF)
and MEK (P-MEK) are shown. Band intensity indicates level of
phosphorylation. Total GAPDH (GAPDH), a housekeeping gene, is used
as a protein loading control. Times, in hours, represent the
duration of compound incubation. NT means “no treatment,” and this
sample serves as a negative control. The level of P-RSK,
highlighted in dotted red rectangles, indicates ERK signaling
activity. The absence of a P-RSK band indicates inhibition of ERK
signaling activity and thereby inhibition of RAS/MAPK pathway
signaling.
In BRAF V600E colorectal cell lines, ERAS-007 also blocked the
RAS/MAPK pathway feedback reactivation observed with MEK or other
ERK plus BRAF inhibitor combinations at one-tenth the concentration
used for the MEK and other ERK inhibitors. These results provide
further support that inhibition of ERK by ERAS-007 may lead to more
complete and durable blockade of the RAS/MAPK pathway relative to
other inhibitors of ERK or MEK, either alone or in
combination.
20
Treatment of two BRAF V600E mutant CRC cell lines, RKO and HT-29,
with encorafenib in combination with the MEK inhibitor binimetinib,
the ERK inhibitor ERAS-007, the ERK inhibitor LY3214996, and the
ERK inhibitor ravoxertinib. The Western blot gels depict
phosphorylation of RSK (P-RSK) and ERK (P-ERK). Higher levels of
phosphorylation are depicted by higher (i.e., darker) band
intensity. Total GAPDH protein (GAPDH) serves as a loading control.
ERK signaling activity is represented by the phosphorylation state
of RSK (P-RSK), which is a downstream target of ERK. The column
values indicate the duration of compound incubation of up to 72
hours.
To explore ERAS-007 activity in vivo, we measured tumor growth
inhibition (TGI) in a melanoma patient-derived xenograft (PDX)
model resistant to BRAF and MEK inhibitors. Whereas treatment with
dabrafenib, a BRAF inhibitor, and vehicle showed similar tumor
growth trajectories, ERAS-007 significantly inhibited tumor growth
at the end of the 38-day treatment period at both 25 mg/kg twice a
day (BID) (p-value < 0.001) and 50 mg/kg BID (p-value <
0.01). These data suggest ERAS-007 may be more potent than BRAF or
MEK inhibitors in achieving inhibition of the RAS/MAPK pathway and
may be able to overcome treatment resistance.
A p-value is the probability that the reported result was achieved
purely by chance, such that a p-value of less than or equal to 0.05
means that there is a less than or equal to 5% probability that the
difference between the control group and the treatment group is
purely due to chance. A p-value of 0.05 or less typically
represents a statistically significant result. The FDA’s
evidentiary standard of efficacy when evaluating the results of a
clinical trial generally relies on a p-value of less than or equal
to 0.05.
21
ERAS-007 shows significant TGI relative to vehicle control at both
25 mg/kg BID (p-value < 0.001) and 50 mg/kg BID (p-value <
0.01) in a dabrafenib resistant melanoma PDX model.
Preclinical anti-tumor activity of ERAS-007
We further showed the breadth of ERAS-007 in vivo activity in CRC,
NSCLC, pancreatic cancer, melanoma, and neuroblastoma models
harboring alterations in the BRAF, NRAS, or KRAS nodes of the
RAS/MAPK pathway. In the BRAF V600E mutant melanoma cell
line-derived xenograft (CDX) A375 model, ERAS-007 showed
dose-dependent tumor inhibition with a maximal 104% TGI at 30 mg/kg
BID (p-value <0.001 across all ERAS-007 doses relative to
vehicle control). In the NRAS Q61K mutant neuroblastoma CDX SK-N-AS
model, ERAS-007 showed dose-dependent tumor inhibition with a
maximal 82% TGI at 25 mg/kg BID (p-value < 0.001 across all
ERAS-007 doses relative to vehicle control). In the KRAS G12D
pancreatic CDX Panc-1 model, ERAS-007 showed dose-dependent TGI
with a maximal 94% TGI at 40 mg/kg BID (p-value < 0.001 across
all ERAS-007 doses relative to vehicle control). In the KRAS G13D
CRC CDX HCT116 model, ERAS-007 showed 101% TGI at 25 mg/kg BID
(p-value < 0.001 relative to vehicle control). ERAS-007 showed
superior TGI to ulixertinib at 75 mg/kg QD in Panc-1 and MIA PaCa-2
at doses ranging from 20 mg/kg BID to 40 mg/kg BID. TGI values
>100% indicated tumor regression.
22
ERAS-007 showed significant TGI in pancreatic cancer, CRC,
melanoma, and neuroblastoma CDX models at doses ranging from as low
as 10 mg/kg BID (p-value < 0.001). At doses ranging from 20
mg/kg BID to 40 mg/kg BID, ERAS-007 showed superior TGI to a
clinical-stage ERK inhibitor, ulixertinib, at 75 mg/kg QD, in
pancreatic cancer Panc-1 and MIA PaCa-2 CDX models. ERAS-007 at 25
mg/kg BID also showed superior TGI to ravoxertinib at 50 mg/kg BID
in the CRC HCT-116 CDX model. Relative to trametinib at 1.5 mg/kg
QD, ERAS-007 showed superior TGI in the MIA PaCA-2 CDX model at
doses ranging from 20 mg/kg BID to 40 mg/kg BID and in the
pancreatic cancer CDX Panc-1 at 40 mg/kg BID. In the neuroblastoma
S-K-NAS model, ERAS-007 showed superior TGI at doses as low as 12.5
mg/kg BID to trametinib at 0.15 mg/kg QD. Error bars represent
standard error of the mean (SEM).
23
ERAS-007 showed statistically significant TGI in BRAF V600E CRC,
mutant EGFR NSCLC, and mutant KRAS NSCLC CDX models as a
monotherapy and in combination with standard of care targeted
therapies and with ERAS-601 (our first MAPKlamp). In the BRAF V600E
CRC CDX model RKO, ERAS-007 exhibited 82% TGI as a monotherapy
(p-value < 0.001), 88% TGI in combination with encorafenib
(p-value <0.001) and 93% TGI in combination with encorafenib and
cetuximab (p-value < 0.001). In the BRAF V600E CRC CDX model
WiDr, ERAS-007 exhibited 102% TGI as a monotherapy (p-value <
0.001), 109% TGI in combination with encorafenib (p-value <
0.001), and 111% TGI in combination with encorafenib and cetuximab
(p-value < 0.001). Indicated with an asterisk in the graphic,
both ERAS-007 combinations achieved statistically significant TGI
relative to either the encorafenib and cetuximab combination or
ERAS-007 monotherapy (p-values < 0.01). In the EGFR exon 19
deletion and MET amplified CDX HCC827/ER1, ERAS-007 achieved 121%
TGI as a monotherapy (p-value < 0.001), 123% in combination with
osimertinib at 10 mg/kg BID (p-value <0.001), and 124% TGI in
combination with osimertinib at 30 mg/kg QD (p-value < 0.001).
In the EGFR exon 19 deletion, EGFR T790M, and MET amplified CDX
NCI-H820, ERAS-007 achieved 117% TGI as monotherapy (p-value <
0.001), 107% TGI in combination with osimertinib at 10 mg/kg BID
(p-value < 0.001), and 112% TGI in combination with osimertinib
at 30 mg/kg QD (p-value < 0.001).
In the KRAS G12V NSCLC CDX model NCI-H441, the MAPKlamp combination
of ERAS-007 at 30 mg/kg QD and ERAS-601 at 15 mg/kg QD achieved a
statistically significant TGI of 113% (p-value < 0.001),
demonstrating statistically significant benefit relative to the
respective monotherapy doses of both ERAS-007 at 30 mg/kg QD and
ERAS-601 at 15 mg/kg QD (p-value < 0.01). ERAS-007 as a
monotherapy at 30 mg/kg BID and 30 mg/kg QD doses achieved
statistically significant TGI of 115% (p-value < 0.001) and 94%
(p-value < 0.001), respectively. ERAS-601 as a monotherapy at 30
mg/kg QD and 15 mg/kg QD doses achieved statistically significant
TGI of 101% (p-value < 0.001) and 87% (p-value < 0.001),
respectively. In the KRAS G12A NSCLC CDX model NCI-H2009, the
MAPKlamp combination of ERAS-007 at 30 mg/kg QD and ERAS-601 at 15
mg/kg QD achieved statistically significant TGI of 107% (p-value
< 0.001). MAPKlamp achieved a statistically significant
combination benefit relative to the respective monotherapy doses of
both ERAS-007 at 30 mg/kg QD and ERAS-601 at 15 mg/kg QD (p-value
< 0.01). The MAPKlamp combination also showed statistically
significant superior TGI relative to ERAS-007 monotherapy at 30
mg/kg BID (p-value < 0.05) and ERAS-601 monotherapy at 30 mg/kg
QD (p-value < 0.01). These doses represent the maximum
monotherapy nonclinical efficacious doses for ERAS-007 and
ERAS-601. ERAS-007 as a monotherapy at 30 mg/kg BID achieved
statistically significant TGI of 93%). ERAS-601 as a monotherapy at
30 mg/kg QD and 15 mg/kg QD doses achieved statistically
significant TGI of 90% (p-value < 0.001) and 73% (p-value <
0.001), respectively. TGI values >100% indicated tumor
regression.
24
ERAS-007 was profiled in two BRAF V600E CRC CDX models, RKO, which
was insensitive to encorafenib and cetuximab treatment, and WiDr,
which was sensitive to encorafenib and cetuximab treatment. In both
models, ERAS-007 combinations showed superior TGI to encorafenib
(Encor.) and cetuximab (Cetux.) monotherapies and to the
encorafenib and cetuximab combination (p-value < 0.01). The
asterisk in the WiDr graphic indicates that the TGI of the ERAS-007
combinations relative to either the encorafenib and cetuximab
combination or ERAS-007 monotherapy was statistically significant
(p-value < 0.01). In two osimertinib-resistant mutant EGFR NSCLC
CDX models, HCC827/ER1 and NCI-H820, ERAS- 007 showed superior TGI
to osimertinib monotherapy both as a single agent and in
combination with osimertinib (p-value < 0.001). In two mutant
KRAS NSCLC CDX models, NCI-H441 and NCI-H2009, the MAPKlamp
combination of ERAS-007 and ERAS-601 achieved statistically
significant TGI relative to vehicle (p-values < 0.01) and showed
statistically significant combination benefit relative to the
respective monotherapy doses used in the MAPKlamp combination
(p-values < 0.01). At their efficacious monotherapy doses,
ERAS-007 and ERAS-601 also achieved significant TGI in both models
as monotherapies (p-values < 0.01).
25
In a panel of 41 CRC PDX models, which include the most common
genetic alterations in CRC, ERAS-007 inhibited tumor growth by
greater than 50% relative to untreated tumors in 30 out of the 41
CRC PDX models (73%). ERAS-007’s inhibitory activity was observed
in 71% of KRAS mutant models (n=17 total), 73% of BRAF models (n=11
total), and 77% of KRAS wildtype and BRAF wildtype models (n=13
total). Together, these in vivo results suggest ERAS-007 exhibits
strong single agent anti-tumor activity across a wide range of
tumors with alterations in BRAF and KRAS relative to other agents
in use today.
ERAS-007 inhibited tumor growth by greater than 50% in 73% of CRC
PDX tumor models. The T/C (treated/control) ratio is calculated as
the ratio of mean volume of ERAS-007 treated tumors to mean volume
of untreated tumors in the control group. Lower values represent
better TGI.
Phase 1 trial of single agent ERAS-007 in patients with advanced
solid tumors
Asana completed a Phase 1 first-in-human clinical trial
(ASN007-101) that evaluated the safety, tolerability, PK,
pharmacodynamics (PD), and preliminary anti-tumor activity of
ERAS-007 in patients with advanced cancers. Forty-nine patients
were enrolled and administered ERAS-007 QD (17 patients) or QW (32
patients). Following dose escalation using both schedules, the
recommended dose (RD) of 250 mg QW was selected. The maximum
tolerated dose (MTD) on a daily schedule was 40 mg QD.
Phase 1 safety and tolerability
ERAS-007 showed a reversible and manageable adverse event profile,
consistent with other RAS/MAPK-pathway inhibitors (e.g., MEK
inhibitors). The ERAS-007 QW dosing schedule was better tolerated
than QD dosing based on the treatment-related adverse events
(TRAEs) reported. Transient nausea and vomiting observed with QW
dosing were manageable. Skin toxicities have been noted as a class
effect of inhibitors of RAF, MEK, or ERK. Less skin toxicity was
observed with QW dosing of ERAS-007 compared to QD dosing.
Ophthalmic toxicities have been observed during treatment with MEK
targeted agents and occur with ERK inhibitors, and reversible
retinopathy is a well-known MEK/ERK inhibitor class
effect.
The ERAS-007 250 mg QW RD was well tolerated with minimal grade 3
and no grade 4 or 5 TRAEs, as shown in the table below. No grade 3
or higher eye toxicity was observed at this dose. We believe these
safety results and QW dosing support combination strategies in our
ERAS-007 clinical development plan.
26
Phase 1 pharmacokinetics
As shown in the table below, the ERAS-007 Phase 1 data suggest that
intermittent QW dosing is preferable to QD dosing. The duration of
time in which mean concentration was above IC90 (%T>IC90), which
correlates with tumor cell killing, was substantially longer for
250 mg QW (~29%) compared to 40 mg QD (~17%). The lower
Cmin
for 250 mg QW, compared to 40 mg QD, allows more time for RAS/MAPK
pathway recovery, which gives normal cells a treatment break during
each dosing interval.
27
Optimizing dosing and scheduling
The observed long biochemical residence time of ERAS-007 bound to
ERK (greater than 9 hours) and the observed human half-life
(approximately 30 hours) offer flexibility in optimizing the dosing
schedule. While the QW schedule of ERAS-007 demonstrated clinical
activity with an acceptable adverse event profile, our exploratory
PK/PD analyses suggest that increasing duration of exposure above
IC90 (which increases tumor cell killing) may drive anti-tumor
activity, and that maintaining Cmin
near or below IC50 (which allows normal cells to recover) may
improve safety and tolerability. Informed by the clinical data
observed from the Phase 1 trial, we conducted PK simulations
(projections based on the data) to explore alternative dosing
regimens to provide additional flexibility for combinations with
ERAS-601 (together, our first MAPKlamp) and other agents, as shown
in the figure and table below.
The PK simulations suggest that the dosing regimen of twice daily
on day 1 of each week (BID-QW) may potentially provide a meaningful
extension of the duration of the PK exposure above the IC90 beyond
what has been achieved with 250 mg QW to improve cancer cell
killing, while still maintaining Cmin
near or below the IC50, to give normal cells a treatment break
during each dosing interval. Therefore, in the Phase 1b/2 trial of
ERAS-007 in patients with advanced or metastatic cancers
(HERKULES-1, described below), we plan to evaluate this BID-QW
dosing schedule in the dose escalation cohort, in addition to the
250 mg QW expansion cohort.
Phase 1 clinical activity
Objective tumor responses and durable disease control of ERAS-007
were observed in diverse tumor types at doses ranging from 120 to
250 mg QW in patients with BRAF-, HRAS-, and NRAS-driven cancers.
As of the December 21, 2020 data cutoff, the waterfall plot below
illustrates the objective responses seen in all patients who had
received at least one dose of ERAS-007 and underwent at least one
tumor assessment (the efficacy-evaluable patients). Four responses
were observed in patients with BRAF V600E thyroid cancer (180 mg
QW), BRAF K601E NSCLC (120 mg QW), HRAS salivary gland cancer (250
mg QW), and BRAF rearranged melanoma (250 mg QW).
28
“% Change from Baseline in Tumor Size” is defined as the maximum
reduction in the sum of tumor diameters of selected target lesions
measured after a patient started on study treatment compared to the
baseline measurements of the same target lesions before study
treatment was started. “Efficacy-Evaluable” is defined as patients
who have a baseline tumor scan and at least one post-baseline scan
after starting study treatment, to allow assessment of tumor
response. Prior treatments in patients with objective responses
included: HRAS salivary gland—radiation; BRAF rearranged
melanoma—nivolumab/ipilimumab, radiation; BRAF V600E
thyroid—radiation; BRAF K601E NSCLC—carboplatin/pemetrexed,
carboplatin/paclitaxel plus durvalumab. The dashed lines represent
the RECIST-defined definitions of response (30% decrease) or
progression (20% increase) in tumor size.
As of the December 21, 2020 data cutoff, the swimmer plot below
demonstrated encouraging duration of treatment, with three patients
who achieved disease control that exceeded one year. One patient
with BRAF V600E melanoma (7104-010) received ERAS-007 for a total
of 71 weeks before experiencing disease progression, first on the
ASN007-101 clinical trial (shown in this swimmer plot) and
subsequently on a single patient IND (not shown in this swimmer
plot), since the patient was deriving clinical benefit at the time
the ASN007-101 trial had completed.
Examples of single agent activity of ERAS-007 from these responses
in patients with melanoma and salivary gland adenocarcinoma are
shown in the scan images below. On the left, a patient with a BRAF
rearranged melanoma, who had prior progression on
nivolumab/ipilimumab, showed complete regression of the target
lesion. On the right, a patient with HRAS salivary gland cancer
showed partial response of target lesions.
29
Rationale for combining with other targeted agents
Since ERK is the terminal node of the RAS/MAPK pathway and
activates hundreds to thousands of downstream proteins, we believe
an ERK inhibitor is an attractive combination partner to achieve
maximal inhibition of the RAS/MAPK pathway. In combination with
RTK, SHP2, RAS, and/or RAF inhibitors, an ERK inhibitor has the
potential to further inhibit RAS/MAPK pathway signaling and delay
development of resistance. The RAS/MAPK pathway is regulated by
negative feedback mechanisms that desensitize the pathway when
active. In the presence of a RAS/MAPK pathway inhibitor, pathway
signaling activity is reduced, alleviating negative feedback
mechanisms and sensitizing the RAS/MAPK pathway to upstream
signaling. This sensitization can prevent RAS/MAPK pathway
inhibitors from achieving therapeutic levels of pathway inhibition.
Another challenge for RAS/MAPK pathway inhibitors is the activation
of RTKs that can generate sufficient upstream RAS/MAPK pathway
signaling pressure that overwhelms RAS/MAPK pathway inhibitors.
Combining upstream RAS/MAPK pathway inhibitors with an ERK
inhibitor can potentially enable pathway inhibition in the absence
of negative feedback and in the presence of additional upstream
signaling pressure. The activity of RAS/MAPK pathway inhibitors can
also be bypassed by the emergence of activating mutations in
RAS/MAPK pathway proteins that lie downstream. For example,
activating mutations in RAS can emerge as a resistance mechanism
against EGFR inhibitors in mutant EGFR NSCLC, and MEK mutations can
develop as a resistance mechanism against BRAF plus MEK inhibitors
in melanoma. As the terminal node of the RAS/MAPK pathway, ERK
inhibition can help address activating RAS, RAF, or MEK mutations
that can act as resistance mechanisms to RAS/MAPK pathway
inhibitors.
BRAF V600E CRC as an example that ERK inhibition can reduce the
emergence of resistance
While the combination of a BRAF inhibitor and an EGFR inhibitor
(encorafenib plus cetuximab) has been approved for the second- and
third-line treatment of BRAF V600E CRC, only 20% of patients
experience an objective response, and only half of these responses
last more than 4 months. Therefore, emergence of resistance is a
major therapeutic barrier to long-term clinical benefit. Analysis
of post-progression biopsies and cell-free DNA samples revealed a
heterogeneous collection of resistance mutations in the RAS/MAPK
pathway, including KRAS, NRAS, MEK1, and MEK2. A set of published
experiments conducted by researchers at Massachusetts General
Hospital modeled this clinical resistance in a pooled clone model
system and xenograft models. Seven different resistant BRAF V600E
CRC cells, each engineered with one of these resistance mutations,
were introduced at 1% allele frequency into a pool of sensitive
BRAF V600E CRC cells. Of all combination therapies evaluated, a
triple blockade of BRAF, EGFR, and ERK (identified with a red box
around the image below) proved to be the most effective in reducing
tumor volume and preventing the emergence of resistance
clones.
30
These data suggest that: (1) in tumors that are highly addicted to
the RAS/MAPK pathway, such as BRAF V600E CRC, resistance mechanisms
are dominated by reactivation of this critical pathway via
mutations within the pathway, and (2) an ERK inhibitor can
potentially overcome these resistance mechanisms by blocking the
terminal node of the pathway. Therefore, ERAS-007 may be combined
with other RAS/MAPK pathway inhibitors (e.g., KRAS G12C inhibitor
and BRAF inhibitor) as either initial therapy or in the
post-progression setting in patients who have been treated with
RAS/MAPK pathway inhibitors.
Development strategy for ERAS-007
We are pursuing a broad clinical development plan for ERAS-007
across multiple tumor types that includes both monotherapy and
combinations with approved and investigational agents. The first
series of trials are four POC trials in solid tumors, NSCLC, CRC,
and AML. While providing POC data, these trials may be expanded to
enable potential accelerated approvals in their respective
indications:
1.
HERKULES-1
for a potential tissue agnostic indication in solid tumors with
RAS/MAPK pathway alterations
2.
HERKULES-2
for EGFR mutant or KRAS mutant NSCLC, with potential to
subsequently expand to other NSCLC populations
3.
HERKULES-3
for KRAS mutant, NRAS mutant, or BRAF V600E CRC initially, with
potential to subsequently expand to other GI cancer
populations
4.
HERKULES-4
for FLT3 mutant AML, with potential to subsequently expand to other
hematological malignancy populations
31
We anticipate multiple data readouts from our HERKULES clinical
trials beginning in 2022, including a Phase 1b monotherapy data
readout from HERKULES-1 in the second half of 2022, a Phase 1b
combination data readout from HERKULES-2 in 2023, and a Phase 1b
combination data readout from HERKULES-3 between the fourth quarter
of 2022 and the first half of 2023.
As shown in the schema below,
HERKULES-1
is a Phase 1b/2 trial evaluating ERAS-007 monotherapy in patients
with solid tumors. In
Part
A,
a dose-finding portion will determine the maximum tolerated dose
(MTD) and RD when ERAS-007 is given BID-QW, the rationale of which
was described earlier in the Phase 1 PK pharmacokinetics section.
The primary endpoint of this part is to characterize the safety
profile of ERAS-007 when given on a BID-QW schedule. Testing this
alternative intermittent dosing schedule will provide us with three
schedules (QD, QW, and BID-QW) from which to select the optimal
dose and schedule to combine with ERAS-601 (our first MAPKlamp) and
other agents. When the ERAS-601 RD is identified from the FLAGSHP-1
trial (described below), we expect to amend the HERKULES-1 trial
protocol to identify the optimal dose and schedule for the ERAS-007
and ERAS-601 combination and to evaluate the preliminary efficacy
and safety of this MAPKlamp in multiple tumor types. In
Part B,
which is running in parallel with Part A (since monotherapy
responses were observed with the QW schedule in the completed Phase
1 clinical trial of ERAS-007), separate cohorts are evaluating
ERAS-007 QW in patients with NSCLC, pancreatic cancer, melanoma,
and other solid tumors that harbor RAS/MAPK pathway alterations.
These patient populations have high unmet medical needs, as they
have exhausted all approved therapies. Patients in some cohorts
have not been previously treated with any RAS/MAPK pathway
inhibitors (e.g., KRAS, BRAF, or MEK inhibitors), since none of
these inhibitors are approved for use in these patients. Patients
in other cohorts have been previously treated with RAS/MAPK pathway
inhibitors when these agents are part of standard of care. The
primary endpoint is an assessment of anti-tumor activity of
ERAS-007 in the patient populations. We dosed the first patient
in
HERKULES-1
in May 2021.
As shown in the schema below,
HERKULES-2
is a Phase 1b/2 master protocol evaluating novel combination
therapies for patients with NSCLC. Sub-Study A is focused on
patients with EGFR mutant NSCLC, representing approximately 184,000
new patients worldwide each year, and Sub-Study B is focused on
patients with KRAS G12C mutant NSCLC, representing approximately
240,000 new patients worldwide each year. The master protocol for
this clinical trial may be expanded in the future to include other
novel combinations and indications in NSCLC. We dosed the first
patient in
HERKULES-2
in September 2021.
Sub-Study A.
For patients with EGFR mutant NSCLC, the standard of care for newly
diagnosed patients with metastatic disease is osimertinib, an EGFR
inhibitor. While 77% of patients respond initially, nearly all
patients experience disease progression while on osimertinib
treatment, and no other targeted therapy is approved in the
post-osimertinib setting. Biomarker analyses of tumors that
developed resistance to osimertinib showed that RAS/MAPK pathway
alterations make up a substantial portion of resistance mechanisms.
With the ERAS-007 plus osimertinib combination, we are evaluating
in this trial whether combined ERK and EGFR inhibition can overcome
osimertinib resistance in EGFR mutant NSCLC with RAS/MAPK pathway
alterations. In
Part A1,
an ERAS-007 QW RD will be identified in combination with
osimertinib. The primary endpoint will be a safety assessment of
this combination. In
Part A2,
this combination regimen will be evaluated in EGFR mutant NSCLC
patients who have progressed on initial osimertinib monotherapy.
The primary endpoint will be an assessment of anti-tumor
activity.
Part A3
will identify the RD of ERAS-007 on a BID-QW schedule in
combination with osimertinib, with a safety assessment as the
primary endpoint.
Part A4
will evaluate this combination with BID-QW schedule in a larger
patient population, with a preliminary assessment of anti-tumor
activity as the primary endpoint.
32
HERKULES-2
Sub-Study A: EGFR mutant NSCLC
Sub-Study B.
For patients with KRAS G12C NSCLC, the standard of care for
previously-treated patients with metastatic disease is sotorasib, a
KRAS G12C inhibitor. Only 36% of patients respond initially, and
nearly all patients experience disease progression. Biomarker
analyses of tumors that developed resistance to a KRAS G12C
inhibitor showed that RAS/MAPK pathway alterations make up a
substantial portion of resistance mechanisms, highlighting the
strong reliance of KRAS G12C NSCLC on this pathway. With the
ERAS-007 plus sotorasib and ERAS-601 plus sotorasib combinations in
Sub-Study B, we are evaluating whether combined ERK and KRAS G12C
inhibition can lead to broader and deeper responses than sotorasib
monotherapy, as well as prolonged efficacy and SHP2 and KRAS G12C
inhibition. In
Part B1 and Part B3,
the RDs for ERAS-007 on QW and BID-QW schedules will be identified
in combination with sotorasib, respectively. The primary endpoint
will be a safety assessment of combinations. In
Part B2 and Part B4,
these combination regimens will be evaluated in KRAS G12C mutant
NSCLC patients who are naïve to KRAS G12C inhibitor treatment. The
primary endpoint will be an assessment of anti-tumor
activity.
Part B5 and Part B7
will identify the RDs of ERAS-601 dosing schedules in combination
with sotorasib, respectively. The primary endpoint will be a safety
assessment of the combinations.
Part B6 and Part B8
will evaluate these combinations in a larger patient population,
respectively, with a preliminary assessment of anti-tumor activity
as the primary endpoint.
HERKULES-2
Sub-Study B: KRAS G12C NSCLC
As shown in the schema below,
HERKULES-3
is a Phase 1b/2 master protocol evaluating novel combination
therapies for patients with GI malignancies. Sub-Study A is focused
on patients with BRAF V600E mutant CRC, representing approximately
180,000 new patients worldwide each year, and Sub-Study B, on
patients with KRAS or NRAS mutant CRC and KRAS mutant pancreatic
cancer, representing over 1.1 million new patients worldwide each
year. The master protocol for this clinical trial may be expanded
in the future to include other novel combinations and indications
in GI cancers. We dosed the first patient in
HERKULES-3
in September 2021.
33
Sub-Study A.
The standard of care for patients with BRAF V600E CRC in the
second-/third-line metastatic setting is encorafenib plus
cetuximab, an anti-BRAF and anti-EGFR doublet therapy. Only 20% of
patients respond, nearly all patients experience disease
progression, and the median overall survival is less than 9 months.
The prognosis for patients in the post-encorafenib plus cetuximab
setting is worse. In preclinical models of BRAF V600E CRC, the
addition of an ERK inhibitor to BRAF inhibitor plus EGFR inhibitor
substantially enhanced anti-tumor activity and reduced the
development of resistance to BRAF inhibitor plus EGFR inhibitor.
In
Part A1,
the ERAS-007 QW RD will be identified in combination with
encorafenib plus cetuximab, with a safety assessment of the
combination as the primary endpoint. The RD will be evaluated
further in two patient populations: BRAF V600E CRC patients who are
naïve to encorafenib plus cetuximab treatment and who have been
treated with encorafenib plus cetuximab. The primary endpoint will
be an assessment of anti-tumor activity.
Parts A2 and
A3
contain optional cohorts that would allow us to evaluate
alternative dosing schedules of ERAS-007 in combination with
encorafenib and cetuximab, depending on the results from Part
A1.
HERKULES-3
Sub-Study A: BRAF V600E CRC
Sub-Study B.
The standard of care for patients with KRAS or NRAS mutant CRC in
the third-line metastatic setting is typically chemotherapy.
However, when treated with regorafenib (a multi-kinase inhibitor)
or TAS-102 (chemotherapy), less than 5% of patients respond, nearly
all patients experience disease progression, and the median overall
survival is around 6 months. In preclinical models of KRAS or NRAS
mutant CRC, the combined inhibition of cycle and RAS/MAPK pathways
demonstrated substantial anti-tumor activity in these highly
resistant tumor types. In
Part B1 and Part B2,
the ERAS-007 QW RD will be identified in combination with two
different schedules of palbociclib, with a safety assessment of the
combination as the primary endpoint. In
Part B3 and Part B4,
the alternative schedule of ERAS-007 given BID-QW may be evaluated.
One RD and schedule will be evaluated further in previously-treated
patients with KRAS or NRAS mutant CRC. The primary endpoint will be
an assessment of anti-tumor activity.
34
HERKULES-3
Sub-Study B: KRAS or NRAS mutant CRC
As shown in the schema below,
HERKULES-4
is a Phase 1b/2 clinical trial evaluating novel combination
therapies for patients with hematological malignancies. Sub-Study A
is focused on patients with FLT3 mutant AML, which represents
30-40% of all AML, or approximately 61,000 new patients worldwide
each year. The standard of care for patients with
relapsed/refractory AML is gilteritinib, a FLT3 inhibitor. Only 14%
of patients achieve a complete remission, nearly all patients
experience disease progression, and the median overall survival is
less than 10 months. While FLT3 is the most commonly altered gene
in AML, alterations along the entire RAS/MAPK pathway are also
prevalent, including SHP2, KRAS, NRAS, and BRAF, suggesting dual
FLT3 and SHP2 inhibition or FLT3 and ERK inhibition may improve the
efficacy of gilteritinib monotherapy. In
Part A1,
the RD of ERAS-007 in two schedules in combination with
gilteritinib will be identified, with a safety assessment as the
primary endpoint. In
Part A2,
a selected RD and schedule of ERAS-007 will be evaluated further in
an expansion cohort of patients with relapsed/refractory FLT3
mutant AML. The primary endpoint will be an assessment of
anti-tumor activity. In
Part A3,
the RD of ERAS-601 in two schedules in combination with
gilteritinib will be identified, with a safety assessment as the
primary endpoint. A selected RD and schedule of ERAS-601 will be
evaluated further in an expansion cohort of patients with
relapsed/refractory FLT3 mutant AML. The master protocol may be
expanded in the future to include additional sub-studies of other
novel combinations and indications in hematological
malignancies.
These and other trial designs may be modified based on evolving
clinical and nonclinical data, as well as feedback from regulatory
agencies.
35
ERAS-601: our SHP2 inhibitor
ERAS-601 is designed to be a potent and selective oral inhibitor of
SHP2. In preclinical studies, ERAS-601 has demonstrated strong in
vitro potency relative to other SHP2 inhibitors (RMC-4550 and
TNO155) and favorable absorption, distribution, metabolism, and
excretion (ADME) and PK properties, which we believe support its
use in a broad range of combination therapies. ERAS-601 is the
second prong of our first MAPKlamp with ERAS-007. In our
first-in-human trial, FLAGSHP-1, we are evaluating the safety,
tolerability, PK, PD, and preliminary anti-tumor activity of
ERAS-601 in patients with advanced or metastatic solid tumors. We
believe that approximately 4.9 million patients worldwide per year
could benefit from ERAS-601 in combination with other agents,
including ERAS-007.
Preclinical profile of ERAS-601
In a biochemical assay, ERAS-601 potently and selectivity inhibited
full length SHP2 with an IC50 value of 4.6 nM as shown in the table
on the left below. By binding to an allosteric pocket that is
present only in the inactive conformation of SHP2, ERAS-601
inhibited SHP2 activity by stabilizing the protein in the inactive
state. No ERAS-601 activity was observed against 10 other
phosphatases (including SHP1), and ERAS-601 showed no strong
inhibition of any kinase in a 300-kinase panel (i.e., less than 30%
inhibition at 1 µM), demonstrating high selectivity as shown in the
table on the right below.
Biochemical on-target activity of ERAS-601 against SHP2 (left) and
biochemical activity of ERAS-601 in a panel of 12 phosphatases
(right). PTPN11 (SHP2) catalytic domain protein is a truncated form
of SHP2 (246 aa – 593 aa). This truncated form contains a
phosphatase domain and is missing two regulatory domains. The
PTPN11 (SHP2) catalytic domain does not harbor the binding site of
ERAS-601 due to these missing domains, while the PTPN11 (SHP2) full
length protein does harbor ERAS-601’s binding site.
ERAS-601’s activity against SHP2 was shown in a cell-based assay
using a SHP2 inhibitor-sensitive cell line, NCI-H1666, which was
transduced with either wildtype SHP2 or mutant SHP2 (T253M /
Q257L). ERAS-601 inhibited RAS/MAPK pathway signaling, shown by a
decrease of phosphorylated ERK (pERK) relative to total ERK in the
figure below, but ERAS-601 had no effect on RAS/MAPK pathway
signaling in the double mutant SHP2 cell line. These cell data
support that ERAS-601’s cellular activity is due to SHP2 binding
and not due to off-target activity. In vitro cell line screening
revealed potent ERAS-601 activity in EGFR, KRAS, NF1, and class III
BRAF mutant cell lines. Generally, ERAS-601 showed greater activity
in RAS/MAPK pathway mutant cell lines that relied on upstream RTK
signaling, such as EGFR, NF1 loss-of-function, and class III BRAF
mutants. ERAS-601 did not show activity in cell lines that harbored
activating RAS/MAPK pathway activating mutations that were not
dependent on upstream signaling, such as the melanoma BRAF V600E
mutant cell line A375.
36
Western blot of the NCI-H1666 cell line transduced with empty
vector (EV), wildtype SHP2 (WT), and double mutant SHP2 (T253M and
Q257L). The two T253M and Q257L mutations in SHP2 prevent ERAS-601
from binding SHP2 via steric hindrance. ERAS-601 at 1 µM inhibited
pERK in empty vector and SHP2 wildtype transduced cells. ERAS-601
at 1 µM did not inhibit pERK in cells transduced with double mutant
SHP2, thereby suggesting that ERAS-601’s cellular activity is due
to SHP2 binding.
The ADME/PK properties of ERAS-601 have been extensively evaluated
in non-clinical studies. As shown in the table below, ERAS-601
demonstrated favorable physicochemical and PK properties, including
low risk of drug-drug interaction (DDI), negligible CYP enzyme
inhibition, and moderate plasma protein binding. It also showed
high oral bioavailability and low clearance across multiple animal
species. We believe these properties support ERAS-601’s use in a
broad range of combination therapies.
37
Preclinical anti-tumor activity of ERAS-601
As shown in the table below, ERAS-601 significantly inhibited tumor
growth as a monotherapy in 25 in vivo models, including KRAS G12C,
KRAS G12D, KRAS G12V, EGFR, BRAF class I and III, and NF1
loss-of-function mutations. In 21 models, ERAS-601 was well
tolerated and showed significant TGI at QD and BID dose schedules.
In a PK/PD study, ERAS-601 also achieved time and dose-dependent
increases in plasma concentrations and concomitant reductions in
RAS/MAPK pathway signaling, as measured by pERK, in the KRAS G12C
mutant NSCLC xenograft model NCI-H358. Tumor pERK1/2 levels were
reduced by more than 50% when ERAS-601 total plasma concentrations
exceeded or approximated the IC50/fu, which is the in vitro
cellular pERK IC50 unbound fraction in plasma.
ERAS-601 exhibited significant TGI relative to vehicle control
(p-value < 0.05) in 11 KRAS mutant, four EGFR mutant, three BRAF
mutant, four NF1 LOF mutant, and three triple wildtype
(KRAS/NRAS/BRAF wildtype) CDX and PDX models. Significant TGI was
observed at both 30 mg/kg QD and 10 mg/kg BID doses. *p-value <
0.05 **p-value < 0.01 ***p-value < 0.001 (p-values assessed
relative to vehicle control)
38
Preclinical activity of ERAS-601 combination therapies
As shown in the figures below, when combined with KRAS G12C and
EGFR inhibitors, ERAS-601 showed significantly greater TGI than
dosing of these inhibitors as monotherapies. This benefit was
observed in models that harbored mutations both upstream and
downstream of SHP2. These ERAS-601 combinations were generally well
tolerated across the tested models as demonstrated by the minimal
percentage body weight changes observed.
ERAS-601 combined with sotorasib and cetuximab showed significant
TGI in five CDX and PDX models. The ERAS-601 + sotorasib (KRAS G12C
inhibitor) showed significant TGI relative to vehicle control
(p-value < 0.01) and monotherapy arms (p-value < 0.05) in
KRAS G12C mutant CRC PDX and esophageal CDX models. The ERAS-601 +
cetuximab (EGFR inhibitor) combination showed significant TGI in
three triple wildtype (KRAS/NRAS/BRAF
wildtype)
CRC PDX models relative to vehicle control (p-value < 0.01) and
cetuximab monotherapy arms (p-value < 0.001).
39
Development strategy for ERAS-601
Our clinical development plan aims to advance ERAS-601 in
combination with other targeted agents to prevent and overcome
adaptive resistance mechanisms in order to achieve more durable
clinical benefit. In the fourth quarter of 2020, we began
evaluating the safety and tolerability of ERAS-601 in a
first-in-human dose escalation trial in patients with advanced or
metastatic solid tumors in our FLAGSHP-1 trial. The primary
endpoints of this trial are assessments of safety and anti-tumor
activity. After we have identified the monotherapy RD of ERAS-601,
we will evaluate rational combinations with ERAS-601, including
dual SHP2 and EGFR inhibition focused on CRC (cetuximab combination
in FLAGSHP-1), dual SHP2 and KRAS G12C inhibition in NSCLC
(sotorasib combination in HERKULES-2), and dual SHP2 and FLT3
inhibition in AML (gilteritinib combination in HERKULES-4), as well
as dual SHP2 and ERK inhibition with ERAS-007 (our first,
innovative MAPKlamp approach) in our HERKULES series of clinical
trials. Other agents for potential combinations include approved
RTK inhibitors, RAS/MAPK pathway inhibitors, and/or investigational
agents we are developing, such as ERAS-3490. Given the wide range
of cancers that are dependent on SHP2, we believe ERAS-601 could
serve as a backbone for compelling combination therapies to prolong
survival for patients.
We anticipate a Phase 1 monotherapy data readout from our FLAGSHP-1
trial in the second half of 2022 and a Phase 1b combination data
readout in triple wildtype (KRAS/NRAS/BRAF wildtype) CRC between
the fourth quarter of 2022 and the first half of 2023.
ERAS-3490: our CNS-penetrant KRAS G12C inhibitor program
RAS proteins are the most frequently mutated oncoproteins, with
KRAS being the most abundantly expressed RAS isoform. Despite
decades of research focused on KRAS as a target of interest in
oncology, it was generally deemed to be undruggable until 2013,
when Dr. Shokat and his colleagues at UCSF identified a new binding
pocket, the "switch II pocket" (S-IIP), via crystallography
studies. Importantly, they also described the discovery of small
molecules that irreversibly bound to this pocket on KRAS G12C – a
finding that turned an undruggable target into a druggable
one.
This historic discovery spurred multiple companies to develop KRAS
G12C inhibitors, including the recently approved sotorasib and
others that are currently in clinical trials. While single agent
activity to date has been most promising in NSCLC, opportunities
remain for improvement in CNS penetration to be able to address the
propensity of NSCLC to metastasize to the brain. Worldwide, KRAS
G12C mutations affect approximately 350,000 patients with cancer,
with NSCLC comprising two-thirds of these patients, and CRC
one-sixth of these patients. NSCLC has the highest rate of CNS
metastases, and the CNS is a site of progression in approximately
25% to 50% of patients on standard of care therapies. Hence, we
believe a CNS-penetrant KRAS G12C inhibitor, either as a
monotherapy or in combination therapies, would represent an
important advance in maintaining systemic disease control,
prolonging response, and preventing CNS progression. In preclinical
in vivo experiments, an approved KRAS G12C inhibitor and a late
clinical-stage KRAS G12C inhibitor (sotorasib and adagrasib,
respectively, and together, the reference compounds) were poorly
CNS-penetrant, detectable in the CNS at less than 10% of the plasma
level. We have been designing and optimizing KRAS G12C inhibitors
that have shown comparable or superior anti-tumor activity to the
reference compounds and robust ability to cross the blood-brain
barrier (BBB) in order to address this key limitation.
Preclinical profile of ERAS-3490 and other
pre-candidates
Our team discovered a novel scaffold that binds the S-IIP in a
different configuration which allows us to optimize potency while
also enabling high CNS penetration. We have discovered and are
characterizing five KRAS G12C inhibitor pre-candidates based on
this scaffold that have promising potency, selectivity, and
physicochemical properties relative to the reference
compounds.
40
Importantly, all five pre-candidates (ERAS-3490, ERAS-3691,
ERAS-3599, ERAS-3537, and ERAS-3788) have shown attractive
physicochemical properties relative to the reference compounds,
especially with respect to in vitro CNS penetration:
Five CNS-penetrant KRAS G12C pre-candidates showed comparable in
vitro and in vivo PK characteristics and in vitro potency to the
reference compounds. ERAS-3490 and ERAS-3599 exhibited superior
exposure and CNS penetration relative to the reference compounds,
and ERAS-3537 and ERAS-3788 exhibited superior CNS penetration
relative to the reference compounds. A green arrow indicates a
favorable value relative to the reference compounds, an orange
arrow indicates a comparable value, and a red arrow indicates an
inferior value. P-gp substrate ratios were characterized in a P-gp
expressing MDCK cell line. The P-gp substrate ratio for a single
reference compound is shown. Per compound, a P-gp substrate ratio
was calculated by dividing its efflux ratio in absence of a P-gp
inhibitor by its efflux ratio in presence of a P-gp inhibitor.
Compounds with lower P-gp substrate ratios are less likely to have
CNS penetration limited by P-gp mediated efflux. LM stands for
liver microsome and CL stands for clearance. Liver microsome
stability is normalized to hepatic blood flow to better enable
cross-species comparisons. In vitro potency was characterized by
both pERK inhibition and cell viability. Both potency assays used
the RAS Initiative KRAS G12C cell line.
We are aiming to optimize CNS penetration while maintaining
comparable potency and metabolic stability to the reference
compounds. This is a highly challenging balancing act to achieve
because typically, the attributes of a molecule that endow it with
potency against the S-IIP (e.g., hydroxyl group moiety for one of
the reference compounds) are the very properties that compromise
its ability to cross the BBB. Our goal with our CNS-penetrant KRAS
G12C inhibitor discovery program has been to significantly increase
CNS penetration as measured by the rat braintotal/plasmatotal
ratio (RBP, measured in percent). Reference compounds have RBPs
ranging from 1% to 6%. Our pre-candidates have RBPs ranging from
11% to 68% (ERAS-3537 [68%], ERAS-3599 [66%], ERAS-3490 [52%],
ERAS-3691 [13%], and ERAS-3788 [11%]), and therefore have the
potential to demonstrate better CNS penetration in the clinic than
the reference compounds. Our team has significant experience
developing targeted CNS-penetrant compounds, including entrectinib
(ROZLYTREK), an FDA-approved product for ROS1 fusion-positive NSCLC
and NTRK fusion-positive solid tumors, including those in patients
with CNS metastases. Using the rat brain assays, entrectinib has an
RBP of 16% and an absolute value of 80 ng/g rat brain
concentration.
Furthermore, ERAS-3490 and ERAS-3599 are not P-glycoprotein (P-gp)
substrates, meaning they are less likely to be effluxed, or pumped,
from the brain back out into the blood. This feature further
enhances the potential for high CNS penetration of these two
pre-candidates relative to the other three pre-candidates and
reference compounds that are P-gp substrates.
Selection of ERAS-3490 as DevCan
ERAS-3490 was derived from a novel scaffold and potently inhibited
the proliferation of 17 KRAS G12C cell lines with IC50s ranging
from 1.4 nM to 82 nM. These values were comparable to reference
compounds, which showed IC50s ranging from 2 nM to 35 nM in the
same cell line panel.
41
ERAS-3490 was also a highly selective inhibitor of KRAS G12C, which
is supported by ERAS-3490 adducting with only one other peptide
when incubated for 4 hours in the KRAS G12C mutant cell line
NCI-H358. The figure below shows that KRAS G12C was strongly
(x-axis) and significantly depleted (y-axis) when incubated with
ERAS-3490, indicating that it strongly and selectively bound to
KRAS G12C.
ERAS-3490 selectively bound to KRAS G12C protein in cell-based
Cysteine Enrichment Proteome (CEP) assays. This assay quantifies
the covalent binding activity of cysteine-targeting compounds, like
ERAS-3490, after being incubated with NCI-H358 cells for 4 hours at
1 µM. The depletion of cysteine-containing proteins in the compound
treatment sample relative to vehicle treatment indicates that the
compound is covalently binding to that peptide. The x-axis
represents the magnitude of protein depletion, and the y-axis
represents the significance of depletion. Proteins in the shaded
green region are significantly depleted by at least 4-fold (p-value
< 1.0e-4) and proteins in the shaded red region are
significantly enriched by at least 4-fold (p-value < 1.0e-4).
Both compounds were profiled in the KRAS G12C mutant NSCLC cell
line NCI-H358. In the ERAS-3490 study, KRAS G12C and one other
protein, PDS5, were the only two proteins out of 2,602 detected
proteins that were significantly depleted.
In an exploratory intracranial KRAS G12C NSCLC CDX NCI-H1373 model
in panel (A) of the figure below, ERAS-3490 demonstrated that its
superior physicochemical properties and PK profile enabled it to
significantly outperform a reference compound in anti-tumor
activity, both at the initial dose of 100 mg/kg QD for the first
nine days and upon lowering the dose to 30 mg/kg for the remainder
of the study. In panel (B), ERAS-3490 demonstrated comparable to
superior anti-tumor activity at two different doses relative to a
reference compound in a subcutaneous pancreatic cancer model (MIA
PaCa-2).
The PK/PD profile help explain why ERAS-3490 outperforms a
reference compound in the intracranial model with its superior PK
profile, with high oral bioavailability (mean plasma
AUC0-lastof
21,606 ng*h/g vs. 10,065 ng*h/g) and high brain bioavailability
(mean brain
AUC0-last
of 1985 ng*h/g vs. ND) at the 30 mg/kg dose. In these in vivo
studies, ERAS-3490 was well tolerated, as body weights remained
constant across all in vivo studies.
42
(A) ERAS-3490 and a reference compound were dosed in an
intracranially injected and luciferase labeled KRAS G12C mutant CDX
model, NCI-H1373. All compounds were dosed at 100 mg/kg QD on days
1-9 and then at 30 mg/kg QD on days 10-42. ERAS-3490 significantly
inhibited tumor growth relative to vehicle control (p-value <
0.01). (B) ERAS-3490 and a reference compound were dosed in a KRAS
G12C inhibitor sensitive pancreatic cancer CDX, MIA PaCa-2, for 28
days. ERAS-3490 significantly inhibited tumor growth at both 30
mg/kg QD (p-value < 0.01) and 100 mg/kg QD (p-value < 0.001)
relative to vehicle control. ERAS-3490 showed minimal body weight
change at doses up to 100 mg/kg QD, demonstrating good
tolerability.
We believe ERAS-3490 is the only KRAS G12C inhibitor specifically
designed to cross the BBB. We nominated ERAS-3490 as our DevCan in
June 2021 and expect to file an IND in the second half of
2022.
Development strategy for ERAS-3490
The initial development of ERAS-3490 as a monotherapy will be in
KRAS G12C mutant NSCLC. We will evaluate the hypothesis that
improved CNS penetration will enhance clinical activity, broaden
the patient population, prolong response, delay disease
progression, and extend survival. Our deep pipeline within the
RAS/MAPK pathway will allow a number of combination therapies to be
developed in NSCLC and other solid tumors, such as CRC and
pancreatic cancer. One of the initial combinations to be assessed
will be with ERAS-601, as preclinical and clinical data support the
synergistic effect of inhibiting KRAS and SHP2. We intend to
explore additional combinations of ERAS-3490 with other approved
and investigational agents.
Our RAS-GTP franchise
Over 2 million patients annually worldwide are affected by RAS
mutations other than KRAS G12C. Like KRAS G12C, these mutations
hyperactivate RAS/MAPK pathway signaling by diminishing RAS’s
ability to transition from the active to the inactive state. Nearly
700,000 of these 2 million patients are affected by tumors that
harbor KRAS G12D, which is the most prevalent KRAS mutation. This
mutation results in hyperactive RAS/MAPK pathway signaling and is
frequently observed in NSCLC, CRC, endometrial cancer, and
pancreatic cancer. Targeting non-G12C RAS mutations (the focus of
our RAS-GTP franchise, including ERAS-4 and ERAS-2/3) is more
challenging than targeting KRAS G12C because: (1) KRAS G12D and
other non-G12C RAS mutations are more commonly found in the active
RAS-GTP state; (2) non-G12C mutations do not have a mutant-specific
site for irreversible inhibitor binding; and (3) these mutations
alter the conformation dynamics of RAS, hindering the ability of
small molecules to target the same binding site as KRAS G12C
inhibitors, S-IIP.
ERAS-4: our KRAS G12D program
Our ERAS-4 program endeavors to develop small molecules that
potently and selectivity bind KRAS G12D. When bound to KRAS G12D,
these inhibitors will prevent RAS-mediated signaling by locking
KRAS G12D in the inactive GDP-bound state and/or obstructing KRAS
G12D’s ability to bind downstream effector proteins, such as BRAF
and CRAF. We are accelerating advancement of this program by
leveraging our in-house chemistry, biology, and structural biology
expertise gained from working on our RAS-GDP and other RAS-GTP
programs.
43
We have generated molecules with low nanomolar IC50 potency against
KRAS G12D in the biochemical RAS-cRAF binding assay and high
selectivity vs. KRAS wildtype (WT). As shown in the figure below,
ERAS-4057 has strong potency of 10.8 nM, with 66-fold selectivity
vs. KRAS WT. We are optimizing the properties of these molecules
utilizing SBDD and structure-activity relationships while
continuing to focus on generating other highly potent and selective
compounds against KRAS G12D, with the intention to nominate and
advance a DevCan into IND-enabling activities.
ERAS-4057 potently and selectively bound KRAS G12D with an IC50 of
10.8 nM (blue) and KRAS WT, an off-target protein, with an IC50 of
717.3 nM (green). Lower values on the y-axis indicate stronger
inhibition of KRAS-RAF1 Ras binding domain (RBD) binding. Higher
values on the x-axis indicate higher concentrations of ERAS-4057.
In this assay, GDP-bound KRAS was converted to GTP-bound KRAS via
an interaction with SOS1 and GTP-bound KRAS then bound the RAF1 RBD
protein. A compound that inhibited the transition of KRAS from the
GDP-bound to GTP-bound state and/or inhibited the protein-protein
binding of KRAS and RAF1 resulted in a lower fraction of KRAS-RAF1
RBD binding.
ERAS-2/3: our other RAS-GTP programs
Our ERAS-2/3 program is focused on the development of small
molecule inhibitors that target a novel region on RAS called the
"switch II groove" (S-IIG). Unlike the S-IIP, the S-IIG is
accessible in both the GDP-bound and GTP-bound states of RAS,
making it a robust binding region across multiple RAS
mutants.
Dr. Shokat identified a new binding site called S-IIG, as shown in
the figure below. The original S-IIP that was the binding site for
KRAS G12C inhibitors is present in the RAS-GDP state only. When RAS
cycles to the RAS-GTP state, the S-IIP becomes obscured by switch
II. Unlike the S-IIP, the S-IIG is not obscured by switch II, which
enables small molecules to access the S-IIG independently of the
phosphorylation state of the bound guanosine. Therefore, S-IIG is
present in both the RAS-GTP and RAS-GDP states. Disruption of these
switch regions can inhibit RAS signaling since GTP-bound RAS binds
to effector proteins at these switch regions. We entered into an
exclusive worldwide license agreement with UCSF for certain
intellectual property derived from Dr. Shokat’s work related to
RAS-GTP, which guides our ERAS-2/3 programs.
44
The small molecule tool compound binds to the S-IIG region on
GDP-bound KRAS M72C in this surface representation of RAS. The
S-IIG is less obstructed by switch II, and this feature allows
small molecules to bind to the S-IIG independently of the
phosphorylation state of the bound guanosine. Unlike S-IIG, access
to S-IIP is influenced by the phosphorylation state of the bound
guanosine. Most selective KRAS G12C inhibitors in development bind
to the S-IIP. Switch II is flexible in the GDP-bound state,
allowing small molecule inhibitors to access the S-IIP. In the
GTP-bound state, switch II rigidly folds over the S-IIP and
occludes access to the S-IIP, thereby preventing most selective
KRAS G12C inhibitors from accessing the S-IIP. Binding of GDP to
RAS is coordinated by a magnesium ion, shown in green.
Our EGFR franchise
EGFR is a transmembrane protein and member of the ErbB family of
receptor tyrosine kinases (RTKs) that under normal conditions bind
various growth factors to activate cellular signaling to regulate
homeostasis. However, when the receptor is overexpressed,
amplified, and/or mutated, it becomes oncogenic, thereby
contributing to cell survival, proliferation, and
metastasis.
We are developing a differentiated portfolio of programs that
target EGFR, including ERAS-801, our CNS-penetrant small molecule
EGFR inhibitor, and ERAS-12, our EGFR domain II/domain III (D2/D3)
targeting bispecific antibody.
ERAS-801: our CNS-penetrant EGFR inhibitor
EGFR-mediated signaling plays a key role in the growth of many
tumor types. Targeting of wildtype EGFR (wtEGFR) and mutant
variants of EGFR (EGFRm) by small molecules and antibodies has
resulted in improved patient outcomes in NSCLC, CRC, and HNSCC.
However, the ability of these agents to effectively target wtEGFR
and EGFRm in the CNS remains an unmet medical need. For example, in
primary CNS tumors like GBM that have amplification of wtEGFR as
well as expression of a mutation in the extracellular domain, the
most common of which is epidermal growth factor receptor variant
III (EGFRvIII), approved small molecule EGFR inhibitors have not
demonstrated clinical activity.
The lack of clinical activity is likely multifactorial, but we
believe there are two primary reasons why approved EGFR inhibitors
are not effective: (1) the molecules do not penetrate the CNS well,
and (2) the molecules are weak inhibitors of the EGFRvIII mutant
protein as homodimers or heterodimers that include wildtype
EGFR.
ERAS-801 is designed to be a potent, selective, reversible, and
orally available small molecule with both: (1) highly enhanced CNS
penetration (3.7:1 brain:plasma ratio in mice) and (2) the ability
to target both EGFR alterations such as EGFRvIII, the most common
mutant form of EGFR found in GBM, and wtEGFR, which heterodimerizes
with EGFRvIII.
45
High CNS penetration of ERAS-801
As shown below, following administration of a single oral dose of
10 mg/kg in mice, ERAS-801 demonstrated substantially higher brain
concentrations than erlotinib, an approved EGFR
inhibitor:
Brain concentrations and exposures of ERAS-801 and erlotinib in
mice when administered a single 10 mg/kg dose. The x-axis
represents time when brain concentration was assessed post-dose.
The y-axis represents total concentration of compound in
nanomolars.
Whereas approved EGFR inhibitors have suboptimal CNS penetration
for primary brain tumors, as shown below, ERAS-801 showed
substantially higher values of Kpand
Kp,uu
(partition coefficients that measure bound and unbound drug
concentration, respectively) compared to osimertinib, afatinib,
erlotinib, gefitinib, and dacomitinib. The figure below is for
illustrative purposes only and is not a head-to-head comparison.
These data were generated from different studies, and caution
should be exercised when comparing data across studies.
46
Dual targeting of EGFR alterations and wtEGFR in GBM to address
heterodimerization
The most common mutant form of EGFR found in GBM is EGFRvIII. Given
the promiscuous nature of EGFR signaling, ERAS-801 has been
specifically designed to have activity against both EGFR
alterations such as EGFRvIII and wildtype EGFR, as we believe that
wtEGFR inhibition is critical to impairing the growth of EGFR
altered GBM because of the propensity of wtEGFR to heterodimerize
with EGFRvIII to drive oncogenic signaling, as seen below with
substantial co-expression of EGFRvIII and wtEGFR.
Panel A showed that the EGFR splice variant mutant EGFRvIII may be
expressed in a subset of GBM tumor cells and that it can be
co-expressed with wildtype EGFR. Panel B showed a zoomed in diagram
of a GBM tumor cell membrane that harbors both wildtype EGFR and
EGFRvIII. Wildtype EGFR can homodimerize with another wildtype EGFR
protein or heterodimerize with EGFRvIII, in each case potentially
leading to oncogenic signaling. In panels C and D, an
immunohistochemistry-stained section of GBM tumor tissue shows
wildtype EGFR-expressing tumor cells in brown and
EGFRvIII-expressing tumor cells in blue. Regions that are stained
both brown and blue express both wildtype EGFR and EGFRvIII
proteins while regions that are stained brown but not blue express
wildtype EGFR only.
Preclinical profile of ERAS-801
In preclinical studies, ERAS-801 has demonstrated strong
biochemical and cell-based potency, as well as strong biochemical
selectivity. ERAS-801 has shown high potency against EGFR with a
biochemical IC50 of 0.3 nM and high CNS penetration. It also showed
high selectivity for EGFR based on a biochemical screen of 484
kinases in which ERAS-801 at 10 µM inhibited only two non-EGFR
family kinases at greater than 90%.
47
The biochemical activities of ERAS-801 and two approved compounds
that are active against EGFR, erlotinib and lapatinib, were
characterized at a single concentration (10 µM) in kinome screens.
Inhibitory activity has been mapped onto a kinase phylogenetic tree
where related kinases within 8 kinase groups are grouped by color
(top row). Red circles indicate kinases where inhibitory activity
has been observed; the diameter of the circle represents the
strength of inhibition (i.e., large circles mean greater inhibitory
activity). Activity against atypical and mutant kinases are shown
in the bottom row.
In cell-based assays, ERAS-801 was potent against wildtype EGFR
with an IC50 of 1.1 nM and EGFRvIII with an IC50 of 0.7 nM. In a 31
patient-derived glioma cell panel, ERAS-801 inhibited the growth of
65% of glioma cells with IC50 values less than 3 µM. This glioma
cell panel included the most frequent types of EGFR alterations
observed in GBM: amplification, EGFRvIII, extracellular domain
mutations (e.g., A289V and A289D), and chromosome 7 polysomy.
ERAS-801 showed no activity in normal human astrocytes (i.e., IC50
greater than 25 µM), which is the most common cell type in the
human brain. ERAS-801’s lack of activity against this normal brain
cell type demonstrated that ERAS-801 selectively inhibited EGFR and
that these normal brain cells were not dependent on EGFR
signaling.
ERAS-801 showed broad activity in a panel of 31 patient-derived
glioma cell lines. Demonstrating selectivity, ERAS-801 showed no
activity in normal human astrocyte cells. Lower IC50 values, in
blue, indicated stronger activity. The EGFR mutation status of the
GBM patient-derived cells is indicated by symbols. Mutations
include extracellular domain EGFR mutations and EGFR splice
variants, such as EGFRvIII. Focal amplification indicates a
high-level gain of the chromosomal region that includes the EGFR
gene locus. Polysomy indicates cells that harbor more copies of
chromosome 7, which contained the EGFR gene, than expected in a
normal cell. Two copies of chromosome 7 are expected in normal
cells.
48
In vivo studies showed that total ERAS-801 concentration was
present in the brain by a factor of 3.7x relative to plasma, and
its unbound concentration in the brain was 1.2x higher than in
plasma. This high CNS penetration translated into enhanced in vivo
survival benefit, which was observed in the EGFRvIII mutant
patient-derived glioma model GBM39. In this study, ERAS-801
significantly extended the survival of mice at 10 mg/kg, 25 mg/kg,
and 75 mg/kg doses relative to vehicle control (p-value < 0.05).
Relative to vehicle control, significant survival benefit was
observed in four additional patient-derived glioma models that
harbor EGFRvIII, EGFR amplified, or chromosome 7 polysomy mutations
(p-value < 0.001). ERAS-801 showed significant survival benefit
and TGI in 93% of 14 patient-derived glioma models that harbored
EGFR amplifications +/- EGFRvIII and extracellular domain EGFR
mutations. These preclinical in vivo data highlight ERAS-801’s
potent CNS activity against EGFR mutant GBM, which comprises 40-60%
of all GBM.
|
|
|
|
ERAS-801 plasma and brain concentrations in mice that have been
administered a single dose of ERAS-801 at 10 mg/kg or 25 mg/kg. The
table summarizes the PK profiles shown in the graph.
ERAS-801 showed dose-dependent survival benefit in the in vivo GBM
PDX model GBM39, which harbored the EGFRvIII splice
variant.
49
ERAS-801 showed significant survival benefit in multiple glioma PDX
models that harbored a variety of EGFR mutations.
***p-value < 0.001.
Development strategy for ERAS-801
We believe that ERAS-801 could provide benefit to approximately
125,000 patients with newly diagnosed GBM worldwide per year. GBM
is a difficult-to-treat, aggressive cancer that can occur in the
brain or spinal cord. Current therapy consists primarily of
surgical resection of the tumor, followed by radiation and
chemotherapy. Once GBM recurs, therapeutic options for patients are
limited. EGFR amplifications and mutations are detected in 40-60%
of GBM cases and are generally indicative of poor prognosis. In the
fourth quarter of 2021, our IND was cleared by FDA to proceed with
a Phase 1 clinical trial of ERAS-801. In February 2022, we dosed
the first patient in our THUNDERBBOLT-1 Phase 1 clinical trial in
recurrent GBM that will evaluate the safety, PK, and PD effects of
ERAS-801 as a single agent. Preliminary evaluation of anti-tumor
activity will also be performed in patients who have tumors
harboring alterations in EGFR.
ERAS-12: our EGFR D2/D3 bispecific antibody program
Inhibition of wildtype EGFR signaling mediated by overexpression of
EGFR has shown promise in treating various tumors, including HNSCC
and CRC. In tumors where overexpression of EGFR is thought to be
the primary driver of EGFR signaling, an antibody-based approach is
the most effective way to target the receptor, and approved
antibodies have demonstrated good tolerability as well as activity
by inhibiting EGFR activation and mediating antibody-dependent
cellular cytotoxicity (ADCC), a process by which the antibody
alerts the immune system to attack the bound tumor cell. However,
all approved anti-EGFR antibodies target domain III (D3) only,
which is the inactive conformation of wildtype EGFR, and no
approved antibodies target domain II (D2), which is the active,
ligand binding, conformation of wildtype EGFR. Antibodies targeting
D2 are expected to be more effective when epidermal growth factor
(EGF) or other members of the EGF family are
overexpressed.
We are developing a bispecific antibody that is active against both
the inactive and active conformations of wildtype EGFR, and we
anticipate filing an IND for this program by 2024.
50
Diagram (A) visualizes the EGFR antibody ER-2a binding to the
extracellular domain II of EGFR wildtype (purple), which is
accessible when EGFR is in the active state. EGFR assumes an active
state conformation when its ligand is bound (the bound ligand is
shown in blue). Diagram (B) visualizes the EGFR antibody ER-3a
binding to the extracellular domain III of EGFR wildtype (purple),
which is accessible when EGFR is in the inactive state. In the
rectangle, the portion of ER-2b that recognizes domain II of EGFR
and the portion of ER-3b that recognizes domain III of EGFR are
combined into a bispecific antibody that binds EGFR in both
states.
By binding to EGFR in the active D2 state, our D2/D3 bispecific
antibody can likely better prevent EGFR dimerization and can
potentially achieve higher levels of EGFR inhibition than currently
approved EGFR antibodies. Achieving a higher level of EGFR
inhibition may better control tumor growth and delay the emergence
of resistance mechanisms involving EGFR that spends more time in
the active conformation.
Targeting D2 via the ER-3a/2a and ER-2a antibodies show a
concentration-dependent inhibition of cancer cell
proliferation.
The bispecific antibody ER-3a/ER-2a and EGFR active state-binding
antibody ER-2a inhibited cell growth in FaDu, an HNSCC cell line,
and HCT-8, a CRC cell line, and the NSCLC cell line H1975. FaDu and
HCT-8 expressed wildtype EGFR and H1975 expressed EGFR with two
kinase domain mutations, L858R and T790M. EGFR’s ligand, EGF, was
added to these cells to further stimulate EGFR activity and model
environments where EGF is expressed. As expected, only the two
antibodies that recognized the active state of EGFR, ER-3a/ER-2a,
inhibited the proliferation of all three cell lines, as indicated
by a reduced confluency percentage.
ERAS-5: our ULK program
The ULK1 and ULK2 kinases are key regulators of the metabolic
process known as autophagy. Under physiological conditions, cells
utilize autophagy to recycle cellular components, breaking down
older components that may be malfunctioning due to age and stress
into subunits that are combined to form new components. This
process can act as a survival mechanism during stress, such as
nutrient starvation, by enabling cells to break down non-critical
cellular components to support critical functions. Autophagy can be
upregulated in tumor cells where RAS/MAPK pathway signaling is
inhibited, acting as an escape route mechanism by preventing tumor
cell death.
51
To pursue our therapeutic strategy of targeting escape routes, our
ERAS-5 program is focused on developing potent, selective
inhibitors of ULK1/2 so that we can further boost tumor cell death
in combination with our RAS/MAPK pathway inhibitors. We have
identified a promising ERAS-5 compound that showed strong potency,
target engagement, inhibition of autophagy, and selectivity. In a
biochemical assay, it exhibited IC50s of 2.4 nM and 2.6 nM against
ULK1 and ULK2, respectively. In cell-based assays, this compound
showed an IC50 of 13.4 nM in an ULK1 target engagement assay and an
IC50 of 5.9 nM in an autophagy pathway activity assay that
visualized GFP-labeled LC3 puncta (LC3 proteins localize on
autophagosomes and, when labeled with a fluorophore, can enable
quantification of autophagosomes by microscopy; higher numbers of
autophagosomes in the cell, which are generated during the
autophagy process, indicate higher levels of autophagy). Our
promising ERAS-5 compound also showed biochemical
selectivity—greater than 375x selectivity against TBK1 and greater
than 240x selectivity against AMPK, two off-target kinases that are
commonly inhibited by other ULK inhibitors with published
structures.
We believe that agents targeting the RAS/MAPK pathway could benefit
from combination with an ULK1/2 inhibitor, addressing up to 2.6
million patients with cancer annually worldwide. This includes over
400,000 patients with RAS mutant pancreatic cancer since pancreatic
cancer tumors have upregulated RAS/MAPK pathway signaling and
autophagy may already be upregulated due to these tumors growing in
nutrient poor environments. As shown below, a promising clinical
case report showed that combining a non-specific autophagy
inhibitor, hydroxychloroquine, with a RAS/MAPK pathway inhibitor,
trametinib, meaningfully reduced tumor burden in a patient with
metastatic pancreatic cancer. The patient’s level of CA19-9, a
blood-based marker of overall tumor burden, rapidly decreased upon
initiation of this combination therapy and remained low through 5
months of treatment. This patient had previously progressed on
multiple chemotherapy regimens and an mTOR inhibitor,
everolimus.
In a patient with metastatic pancreatic cancer, the combination of
an autophagy inhibitor, hydroxychloroquine, with a RAS/MAPK pathway
inhibitor, trametinib, resulted in a steep reduction of overall
tumor burden as represented by a decrease in CA19-9. Higher tumor
burden levels are shown as higher concentrations of CA19-9. The
x-axis represents dates within the patient’s treatment journey and
the y-axis is the detected concentration of CA19-9. Prior to
initiation of trametinib and hydroxychloroquine treatment, the
patient was treated with chemotherapy regimens mFOLFIRINOX (folinic
acid [leucovorin], fluorouracil, irinotecan, and oxaliplatin),
gemcitabine, and capecitabine, and gemcitabine, abraxane, and
cisplatin. After progressing on the gemcitabine, abraxane, and
cisplatin triplet, the patient was treated with an mTOR inhibitor,
everolimus. The patient progressed on everolimus and was then
treated with trametinib at 2 mg QD in combination with escalating
doses of hydroxychloroquine up to 1,200 mg QD. The combination of
trametinib at 2 mg QD with hydroxychloroquine at 1,200 mg resulted
in a significant decrease in overall tumor burden that continued
through 5 months of treatment.
52
ERAS-9: our SOS1 program
SOS1 is a protein that binds to RAS and enables it to transition
from the inactive RAS-GDP state to the active RAS-GTP state. RAS
proteins bind GDP tightly, and a cofactor, such as a SOS1, is
required to facilitate RAS’s release of GDP followed by its binding
to GTP. Without this cofactor, RAS will accumulate in the inactive
state as active state RAS hydrolyzes bound GTP. We are developing
small molecule inhibitors in our ERAS-9 program that obstruct
SOS1-RAS binding and thereby prevent RAS from cycling to the active
RAS-GTP state. SOS1-RAS inhibition can prevent RAS activation
mediated by upstream signaling (e.g., via EGFR activation) and can
be combined with downstream RAS/MAPK pathway inhibitors to
potentially address RAS and RAF mutations that result in
constitutive RAS/MAPK pathway signaling. SOS1-RAS inhibitors can
address as many as 4.9 million patients with cancer who harbor
RAS/MAPK pathway activating mutations annually worldwide either as
a monotherapy or in combination therapies.
ERAS-10: our protein degrader program
We are exploring protein degradation as an alternative mechanism to
complement our approach of enzymatically inhibiting oncogenic
proteins. Degrader molecules bind to a target oncogenic protein and
cellular machinery that label proteins for degradation. Within
proximity of each other, the degrader machinery labels the target
protein through a process called ubiquitination, and the labeled
protein is degraded. Degraders can offer advantages over enzymatic
inhibitors, such as the ability of a single degrader molecule to
tag many copies of the target oncoprotein for degradation and the
ability of a degrader to more effectively inhibit the function of
non-enzymatic proteins. We think this approach will allow us to
target a broader range of proteins within the RAS/MAPK pathway and
may help us more effectively target a subset of oncogenic proteins
than via enzymatic inhibition alone.
ERAS-11: our MYC program
MYC is a transcription factor that is mutated in 40% of cancers,
affecting approximately 7.7 million patients with cancer annually
worldwide. These mutations promote cancer by hyperactivating MYC
and/or its protein dimerization partners (e.g., MAX). Inhibiting
MYC by disrupting its ability to dimerize with other proteins or
bind DNA has been pursued for over 20 years but has not yet been
successful. We are exploring novel approaches to targeting MYC
utilizing our internal discovery expertise complemented with
partnerships to overcome the challenges that have prevented the
successful development of MYC protein inhibitors.
Our acquisition and license agreements
Asana BioSciences
In November 2020, we entered into an agreement and plan of merger
with Asana and ASN Product Development, Inc. (ASN) (the Asana
Merger Agreement), pursuant to which ASN became our wholly-owned
subsidiary. Asana and ASN had previously entered into a license
agreement, which was amended and restated prior to the closing of
the merger transaction (the Asana License Agreement, and
collectively with the Asana Merger Agreement, the Asana
Agreements), pursuant to which ASN acquired an exclusive, worldwide
license to certain intellectual property rights relating to
inhibitors of ERK1 and ERK2 owned or controlled by Asana to develop
and commercialize ERAS-007 and certain other related compounds for
all applications. We have the right to sublicense (through multiple
tiers) the licensed rights under the Asana Agreements, subject to
certain conditions. The foregoing license is subject to Asana’s
non-exclusive right to practice the licensed rights to research and
conduct preclinical pharmacology activities with a specified
combination of compounds, subject to certain specified conditions.
Pursuant to the Asana License Agreement, neither Asana nor ASN can
directly or indirectly exploit certain classes of competing
products, subject to specified exceptions. In addition, we are
required to use commercially reasonable efforts to develop and
obtain regulatory approval for ERAS-007 in the United States, at
least one major market country in Europe, and either China or
Japan.
Under the Asana Merger Agreement, we made an upfront payment of $20
million and issued 4,000,000 shares of our Series B-2 convertible
preferred stock to Asana. In connection with our IPO, these shares
of Series B-2 convertible preferred stock were converted into
3,333,333 shares of our common stock. We are obligated to make
future development and regulatory milestone cash payments for a
licensed product in an amount of up to $90 million. Additionally,
upon achieving a development milestone related to demonstration of
successful proof-of-concept in a specified clinical trial, we will
be required to issue 3,888,889 shares of our common stock to Asana.
We are not obligated to pay royalties on the net sales of licensed
products.
53
Upon our payment to Asana of all merger consideration, including
upfront cash and equity payments, the milestone payments, the
equity payment related to the proof-of-concept development
milestone, and all other development milestone payments, with the
exception of a specific milestone that does not need to be achieved
at such time and will remain subject to payment in the event that
such milestone occurs at a later time, all licensed rights will
become fully paid-up, perpetual, and irrevocable. The License
Agreement may be terminated by either Asana or us in the event of
an uncured material breach by the other party. Asana also has the
right to terminate the Asana License Agreement if we fail to engage
in material activities in support of clinical development and
commercialization of ERAS-007 for a period of 12 consecutive
months, excluding reasons outside of our reasonable control and
subject to certain limitations. However, Asana’s right to terminate
the Asana License Agreement for any reason ends once we have paid
to Asana all merger consideration, or if Asana’s equity interest in
us is publicly traded and exceeds a certain threshold value. We may
terminate the Asana License Agreement at any time upon the
provision of prior written notice to Asana.
NiKang Therapeutics
In February 2020, we entered into a license agreement (the NiKang
Agreement) with NiKang Therapeutics, Inc. (NiKang) under which we
were granted an exclusive, worldwide license to certain
intellectual property rights owned or controlled by NiKang related
to certain SHP2 inhibitors to develop and commercialize ERAS-601
and certain other related compounds for all applications. We have
the right to sublicense (through multiple tiers) our rights under
the NiKang Agreement, subject to certain conditions, and are
required to use commercially reasonable efforts to develop and
commercialize licensed products. The parties are obligated to
negotiate in good faith for a certain period of time to grant
NiKang the exclusive commercial distribution rights in greater
China once a licensed product reaches a certain development
stage.
Under the NiKang Agreement, we made an upfront payment of $5
million to NiKang and reimbursed NiKang $0.4 million for certain
initial manufacturing costs. In addition, we paid an additional $7
million after publication of a US patent application that covered
the composition of matter of ERAS-601. We are also obligated to pay
(i) development and regulatory milestone payments in an aggregate
amount of up to $16 million for the first licensed product and $12
million for a second licensed product, and (ii) commercial
milestone payments in an aggregate amount of up to $157 million for
the first licensed product and $151 million for a second licensed
product. We are also obligated to: (i) pay tiered royalties on net
sales of all licensed products in the mid-single digit percentages,
subject to certain reductions; and (ii) equally split net
sublicensing revenues earned under sublicense agreements that we
enter into with any third party before commencement of the first
Phase I clinical trial for a licensed product.
The NiKang Agreement will expire upon the last to expire royalty
term, which is determined on a licensed product-by-licensed product
and country-by-country basis, and is the later of: (i) ten years
from the date of first commercial sale, (ii) the last to expire
valid claim within the licensed patent rights covering such
licensed product, or (iii) the expiration of all regulatory
exclusivity for the licensed product in such country. Upon
expiration of the NiKang Agreement, on a licensed
product-by-licensed product and country-by-country basis, we will
have a fully paid-up, non-exclusive license to conduct research and
to develop and commercialize the licensed products.
The NiKang Agreement may be terminated in its entirety by NiKang in
the event of our uncured material breach, which includes our
failure to use commercially reasonable efforts to satisfy certain
specified clinical development diligence milestones. In addition,
NiKang may terminate if we, directly or indirectly, commence a
legal action challenging the validity or enforceability of any
licensed patents. Further, if we acquire more than 50% of the
equity or assets of a company that owns a competing small molecule
that is designed to prevent the same target as set forth in the
NiKang Agreement from switching to an enzymatically active state,
then we must either divest such competing product or terminate the
NiKang Agreement. We may terminate the NiKang Agreement at any time
upon the provision of prior written notice to NiKang. Upon
termination of the NiKang Agreement for any reason, all rights and
licenses granted to us, as well as any sublicenses that we granted
thereunder, will terminate. In addition, upon any termination (but
not expiration) of the NiKang Agreement and upon NiKang’s request,
the parties are obligated to meet and negotiate in good faith the
terms of a license from us to NiKang to allow NiKang’s continued
development, manufacture, and commercialization of the licensed
products.
54
Katmai Pharmaceuticals
In March 2020, we entered into a license agreement (the Katmai
Agreement) with Katmai Pharmaceuticals, Inc. (Katmai) under which
we were granted an exclusive, worldwide, royalty-bearing license to
certain patent rights and know-how controlled by Katmai related to
the development of small molecule therapeutic and diagnostic
products that modulate EGFR and enable the identification,
diagnosis, selection, treatment, and/or monitoring of patients for
neuro-oncological applications to develop, manufacture, use, and
commercialize ERAS-801 and certain other related compounds in all
fields of use. We have the right to sublicense (through multiple
tiers) our rights under the Katmai Agreement, subject to certain
limitations and conditions, and are required to use commercially
reasonable efforts to develop, manufacture, and commercialize
licensed products and to meet certain specified development and
launch milestones by certain dates. We are obligated to use
commercially reasonable efforts to develop the licensed products
first for use within the neuro-oncology field before expanding our
development efforts to include other indications in the oncology
field. Following the first achievement of a clinical
proof-of-concept for any indication, we have the right to submit a
non-binding offer to Katmai for: (i) the purchase of all licensed
patent rights, know-how, and other assets owned by Katmai that are
necessary or useful for the exploitation of the licensed products,
or (ii) for the purchase of Katmai. Pursuant to the Katmai
Agreement, neither Katmai nor we can directly or indirectly exploit
certain specified classes of competing products.
The license granted under the Katmai Agreement is subject to The
Regents of the University of California’s reserved right to: (i)
use the licensed patent rights and know-how for educational and
non-commercial research purposes, and to publish results arising
therefrom, and (ii) grant licenses to the licensed know-how to
third parties without notice because the licensed know-how is
non-exclusively licensed to Katmai by The Regents of the University
of California. Further, the license granted under the Katmai
Agreement is subject to the rights of the United States government
under the Bayh-Dole Act, including: (i) a non-exclusive,
non-transferable, irrevocable, paid-up license to practice or have
practiced the invention claimed by the licensed patent rights
throughout the world, and (ii) the obligation that any licensed
products used or sold in the United States be manufactured
substantially in the United States.
Under the Katmai Agreement, we made an upfront payment of $5.7
million and Katmai agreed to purchase shares of our Series B-1
convertible preferred stock and Series B-2 convertible preferred
stock having an aggregate value of $2.7 million. In connection with
our IPO, these shares of Series B-1 convertible preferred stock and
Series B-2 convertible preferred stock were converted into 395,555
shares of our common stock, in the aggregate. We are obligated to
make future development and regulatory milestone payments of up to
$26 million and commercial milestone payments of up to $101
million. We are also obligated to pay tiered royalties on net sales
of each licensed product, at rates ranging from the mid- to
high-single digit percentages, subject to a minimum annual royalty
payment in the low six figures and certain permitted
deductions.
Our royalty obligations and the Katmai Agreement will expire, on a
licensed product-by-licensed product and country-by-country basis,
on the earlier of: (i) the ten-year anniversary of the expiration
of all valid claims included in the licensed patents covering the
composition of matter or method of use of such licensed product in
such country, or (ii) the twentieth anniversary of the first
commercial sale of such licensed product in such country. Upon the
expiration of the Katmai Agreement, we will have a fully paid-up
and irrevocable license.
The Katmai Agreement may be terminated in its entirety by either
party: (i) in the event of an uncured material breach by the other
party, or (ii) in the event the other party becomes subject to
specified bankruptcy, insolvency, or similar circumstances.
Provided that we are in full compliance with the Katmai Agreement,
we may terminate the Katmai Agreement upon written notice to
Katmai. Upon termination of the Katmai Agreement for any reason,
all rights and licenses granted to us thereunder will terminate.
Upon termination of the Katmai Agreement, we are obligated, among
other things, to: (i) grant an exclusive license to Katmai under
all of our right, title and interest in all inventions and know-how
developed under the Katmai Agreement existing at the time of
termination that are specific to the licensed compounds or
products, including without limitation all data and results related
to their exploitation, and (ii) transfer to Katmai ownership and
possession of all regulatory filings related to the licensed
compounds and products. Unless the Katmai Agreement is terminated
for our material breach, the parties will negotiate in good faith
the financial terms pursuant to which the foregoing actions will be
conducted, provided that our performance of such actions may not be
conditioned upon the conduct or completion of such negotiations. If
the parties are unable to agree upon such terms within the
specified time period, then the parties will submit all unresolved
matters for resolution by arbitration.
55
Emerge Life Sciences
In March 2021, we entered into an asset purchase agreement (the ELS
Purchase Agreement) with ELS wherein we purchased all rights,
title, and interest (including all patent and other intellectual
property rights) to ELS’s EGFR antibodies directed against the EGFR
domain II (EGFR-D2) and domain III (EGFR-D3) as well as a
bispecific antibody where one arm is directed against EGFR-D2 and
the other is directed against EGFR-D3 (the Antibodies). Under the
ELS Purchase Agreement, we issued to ELS 500,000 shares of our
common stock and made an upfront payment of $2 million. We are not
obligated to pay royalties on the net sales of products covered by
the acquired intellectual property. Under the ELS Purchase
Agreement, ELS is committed to performing certain studies on the
Antibodies to assist in development activities, the costs of which
shall be mutually agreed upon and for which we will be
responsible.
Pursuant to the ELS Purchase Agreement, at any time between 12
months and 36 months after the effective date of the ELS Purchase
Agreement, if we reasonably determine that none of the Antibodies
should be taken into human clinical trials due to safety, efficacy
or CMC issues, then we have the option to select another antibody
developed and solely owned by ELS that is not the subject of a
license, collaboration, or option to a third party (the Option). If
we elect to exercise the Option, then ELS will provide us with a
list of all available antibodies that meet the aforementioned
requirements, and we have the right to select one antibody from the
list. Upon our selection of an antibody, ELS will assign us all
rights, title and interest to such antibody (including patent and
other intellectual property rights) subject to any pre-existing
obligations or restrictions. In the event that we wish to have ELS
conduct any studies on such optioned antibody, then after mutual
agreement as to the scope of the studies, we will be responsible
for the cost for such studies.
LifeArc
In April 2020, we entered into a license agreement with LifeArc
(the LifeArc Agreement) under which we were granted an exclusive,
worldwide license to certain materials, know-how, and intellectual
property rights owned or controlled by LifeArc to develop,
manufacture, use, and commercialize certain ULK inhibitors for all
applications. We also have the right to sublicense (through
multiple tiers) our rights under the LifeArc Agreement, subject to
certain conditions. The foregoing license is subject to LifeArc’s
retained non-exclusive, irrevocable, worldwide, sublicensable (to
its academic collaborators), royalty-free right to use the licensed
intellectual property rights within all fields of use for LifeArc’s
own non-commercial, non-clinical academic research. Notwithstanding
its retained rights, LifeArc will not seek to develop or undertake
any other ULK1/2 therapeutic development programs either in-house
or via third parties until April 2025. We are required to use
diligent efforts to achieve certain development and regulatory
milestones with respect to submission of an IND, initiation of
clinical trials, submission of a new drug application (NDA), and
commencement of commercial sales.
Under the LifeArc Agreement, we were granted the license at no
upfront cost and a period of three months after the effective date
to conduct experiments on LifeArc’s compounds. Upon completion of
this initial testing period, we had the option to continue the
license and make a one-time license payment of $75,000 to LifeArc,
which payment was subsequently made. We are obligated to make
future development milestone payments for a licensed product of up
to $11 million and sales milestone payments of up to $50 million.
We are also obligated to pay royalties on net sales of all licensed
products, in the low-single digit percentages, subject to certain
reductions.
Our royalty obligations and the LifeArc Agreement will expire, on a
licensed product-by-licensed product and country-by-country basis,
on the later of: (i) ten years from the date of first commercial
sale, and (ii) when there is no longer a valid patent claim
covering such licensed product, or expiration of regulatory
exclusivity for the licensed product in such country. Upon
expiration of the LifeArc Agreement, all rights and licenses
granted to us and under the LifeArc Agreement will continue on a
fully paid-up basis.
The LifeArc Agreement may be terminated in its entirety by either
LifeArc or us in: (i) the event of an uncured material breach by
the other party, or (ii) in the event the other party becomes
subject to an order by a court of competent jurisdiction for
winding-up or dissolution or similar circumstances. Further,
LifeArc may terminate the LifeArc Agreement by giving written
notice to us if: (i) we fail to comply with our diligence
obligations and fail to take remedial actions, (ii) we fail to
agree on a mechanism to cure a persistent breach, or (iii) we fail
to provide proof of the insurance coverage as required under the
LifeArc Agreement. We may terminate the agreement at any time upon
the provision of written notice to LifeArc.
56
Upon termination of the LifeArc Agreement for any reason, all
rights and licenses granted to us, as well as any sublicenses we
granted thereunder, will terminate. In addition, upon termination
of the LifeArc Agreement for any reason other than its natural
expiration or termination by us for LifeArc’s material breach,
LifeArc has an option to negotiate an exclusive, worldwide,
sublicensable license to commercialize any patent rights, technical
and clinical data, and any development results relating to the
licensed products that are owned or controlled by us for the
purpose of developing, manufacturing and commercializing the
licensed products on terms to be negotiated between the
parties.
University of California, San Francisco
In December 2018, we entered into a license agreement, as amended
(the UCSF Agreement), with The Regents of the University of
California, San Francisco (the Regents), under which we were
granted an exclusive, worldwide, royalty-bearing license under
certain patent rights claiming novel covalent inhibitors of GTP-
and GDP-bound RAS for the development and commercialization of
products covered by such patent rights for the prevention,
treatment and amelioration of human cancers and other diseases and
conditions. We have the right to sublicense (through multiple
tiers) our rights under the UCSF Agreement, subject to certain
conditions. The UCSF Agreement was amended in May 2021. The
foregoing license is subject to various retained rights and
restrictions, including: (i) the Regents’ reserved right to make,
use and practice the licensed patent rights and any technology
relating thereto for educational and research purposes, (ii) Howard
Hughes Medical Institute’s non-exclusive, fully paid-up,
irrevocable worldwide license to use the licensed patent rights for
research purposes, (iii) Howard Hughes Medical Institute’s
statement of policy on research tools, and (iv) the obligations to
the US government under the Bayh-Dole Act, including the obligation
to report on the utilization of the invention covered by the
licensed patent rights and a non-exclusive, non-transferable,
irrevocable, paid-up license to practice or have practiced such
invention throughout the world. We are required to use diligent
efforts to proceed with the development and commercialization of
licensed products including by achieving certain milestone events
within the specified time periods.
Under the UCSF Agreement, we made upfront payments of $50,000 to
the Regents and pay the Regents an annual license maintenance fee,
but such fee will not be due on any anniversary if, on that date,
we are making royalty payments to the Regents. We are obligated to
make future development and regulatory milestone payments of up to
$6.4 million and a sales milestone payment of $2 million for either
of the first two licensed products. We are also obligated to pay
royalties on net sales of all licensed products in the low-single
digit percentages, subject to a minimum annual royalty payment in
the low six figures, commencing on the year of the first sale of a
licensed product and continuing, on a licensed product-by-licensed
product and country-by-country basis, until there are no valid
claims of the licensed patent rights covering the licensed product
in such country. Additionally, we are obligated to pay tiered
sublicensing fees, with the first two tiers in the low-to-mid teen
percentages and the third tier at 30%, on certain fees we receive
from any sublicense that we grant, depending on the stage of
development of a licensed product when such sublicense is granted.
Prior to the execution of the amendment, we were obligated to make
a cash payment to the Regents in the event of our initial public
offering, a change of control transaction or a reverse merger (the
Corporate Milestone). In the amendment, the amount of the cash
payment payable upon our achievement of a Corporate Milestone was
reduced and we agreed to issue the Regents 944,945 shares of our
common stock, which issuance is not contingent upon the achievement
of a Corporate Milestone and occurred in May 2021. In August 2021,
following the achievement of the Corporate Milestone, we made a
cash payment to the Regents in the amount of $1.7
million.
The UCSF Agreement will expire upon the expiration of the last of
the licensed patent rights. The UCSF Agreement may be terminated in
its entirety by the Regents: (i) for our uncured breach, (ii) for
our bankruptcy, or (iii) if we challenge, directly or indirectly,
the validity or enforceability of any licensed patents. Further, if
we fail to satisfy any diligence milestones, the Regents has the
right and option to either terminate the UCSF Agreement or modify
the exclusive license granted thereunder to a non-exclusive
license. We may terminate the UCSF Agreement in its entirety or on
a country-by-country basis at any time upon the provision of
written notice to the Regents. Upon termination of the UCSF
Agreement for any reason, all rights and licenses granted to us
thereunder will terminate.
57
Commercialization
We intend to maintain exclusive worldwide development and
commercialization rights to our product candidates (excluding
programs in our pipeline that arise from an investment made by
Erasca Ventures in a third party) and, if marketing approval is
obtained, to commence commercialization activities by building a
focused sales and marketing organization to sell our products on
our own in the United States and potentially other regions such as
Europe. We will likely seek commercialization partnerships for our
product candidates in other regions beyond the United States and
Europe. We currently have no sales, marketing, or commercial
product distribution capabilities. We intend to build the necessary
infrastructure and capabilities over time for commercialization in
the United States and potentially other regions, following further
advancement of our product candidates. Clinical data, the size of
the addressable patient population, the size of the commercial
infrastructure and manufacturing needs, and the status of our
pipeline, may all influence or alter our commercialization
plans.
Competition + Cooperation (“Coopetition”)
Although the biotechnology and pharmaceutical industries, and the
oncology sector, are characterized by rapid evolution of
technologies, fierce competition, and strong defense of
intellectual property rights, we believe the most fearsome
competitor of all is cancer itself. As such, we view other
companies in this sector more as potential allies and collaborators
than as competitors, as we all have a common cause: to defeat
cancer. Many of the companies that are developing or marketing
treatments for cancer, including major pharmaceutical and
biotechnology companies that are working on therapies targeting the
RAS/MAPK pathway, are companies with whom we endeavor to
collaborate in our mission to erase cancer.
Collaborating with these companies alleviates some of the
traditional challenges that emerging companies face with respect to
financial resources, established presence in the market, expertise
in research and development, manufacturing, preclinical and
clinical testing, obtaining regulatory approvals and reimbursement,
and marketing approved products. Similarly, recruiting and
retaining qualified scientific and management personnel,
establishing clinical trial sites and patient registration for
clinical trials, as well as in acquiring technologies complementary
to, or necessary for, programs are challenges for all companies
developing or marketing treatments for cancer.
That said, our commercial potential could be reduced or eliminated
if other companies develop and commercialize products that are
safer, more effective, have fewer or less severe side effects, are
more convenient or are less expensive than products that we may
develop. Other companies also may obtain FDA or other regulatory
approval for their products more rapidly than we may obtain
approval for ours, which could result in these companies
establishing a strong market position before we are able to enter
the market or make our development more complicated.
There are numerous companies developing or marketing treatments for
cancer, including many major pharmaceutical and biotechnology
companies. These treatments consist of small molecule drug
products, biologics, cell-based therapies, and traditional
chemotherapy. There are also a number of pharmaceutical companies
with product candidates in development that target the nodes
involving the RAS/MAPK pathway. These include, among others, Amgen,
AstraZeneca, Black Diamond Therapeutics, BioMed Valley Discoveries,
Boehringer Ingelheim, Deciphera Pharmaceuticals, Eli Lilly, Jacobio
Pharmaceuticals (in collaboration with AbbVie), Janssen, Merck,
Mirati Therapeutics, Navire Pharma (a subsidiary of BridgeBio),
Novartis, Pfizer, Relay Therapeutics (in collaboration with
Genentech), Revolution Medicines, Roche/Genentech, Sanofi, and
Schrödinger (in collaboration with Bristol Myers
Squibb).
Intellectual property
We strive to protect the proprietary technology, inventions, and
improvements that are commercially or strategically important to
our business, including seeking, maintaining, and defending patent
rights, whether developed internally or in-licensed/acquired from
third parties. We also rely on trade secrets and know-how relating
to our proprietary technology and product candidates and continuing
innovation to develop, strengthen and maintain our proprietary
position. We also plan to rely on data exclusivity, market
exclusivity and patent term adjustments or extensions when
available. Our commercial success will depend in part on our
ability to obtain and maintain patent and other intellectual
property protection for our proprietary technology, inventions and
improvements; to preserve the confidentiality of our trade secrets;
to defend and enforce our proprietary rights, including any patents
or trademarks that we may own in the future; and to operate without
infringing on the valid and enforceable patents and other
proprietary rights of third parties. Intellectual property rights
may not address all potential threats to our competitive
advantage.
58
We continually assess and refine our intellectual property strategy
as we develop new product candidates. To that end, we are prepared
to file additional patent applications in any appropriate fields if
our intellectual property strategy includes such filings, or where
we seek to adapt to competition or seize business opportunities.
Further, we are prepared to file patent applications, as we
consider appropriate under the circumstances, relating to the new
technologies that we develop.
We cannot be sure that patents will be granted with respect to any
of our pending patent applications or with respect to any patent
applications we may own or license in the future, nor can we be
sure that any of our existing patents or any patents we may own or
license in the future will be useful in protecting our
technology.
To cover our proprietary technologies and our current pipeline of
proprietary product candidates and related methods, such as methods
of use, we have issued patents and patent applications representing
thirty-eight patent families. As of December 31, 2021, our patent
estate, which consists of owned and in-licensed patent families,
includes seven issued US patents, eleven pending US non-provisional
patent applications, fifty-one pending US provisional patent
applications, three issued foreign patents, eleven pending
international patent applications filed under the Patent
Cooperation Treaty (PCT application), and ninety-four pending
foreign patent applications in various markets outside of the
United States. In particular, we have patent applications pending
for each of our product candidates.
ERAS-007
As of December 31, 2021, we have in-licensed three patent families
from Asana. The three patent families relate to ERK 1/2 inhibitors,
their preparation, and methods of use. One of the families covers
the ERAS-007 product candidate compound and additional ERK1/2
inhibitor compounds, their preparation and method of use, and
includes four issued US patents, one pending US non-provisional
patent application, three issued foreign patents, and fourteen
pending foreign patent applications. The second and third families
cover methods of using ERAS-007, and includes one pending US
provisional patent application, one pending PCT application and two
pending foreign patent applications. The issued US patents are
expected to expire in June 2036, absent any patent term adjustments
or extensions. Any patents issued from the patent applications
related to ERAS-007 are expected to expire between 2036 and 2042,
absent any patent term adjustments or extensions.
As of December 31, 2021, we also own six patent families relating
to ERAS-007. The patent families include fifteen pending US
provisional patent applications. Any patents issued from these
patent applications are expected to expire in 2042, absent any
patent term adjustments or extensions.
ERAS-601
As of December 31, 2021, we have in-licensed two patent families
from NiKang. The two patent families relate to SHP2 inhibitor
compositions, their preparation, and methods of use. One of the
families covers the ERAS-601 product candidate compound, its
preparation and method of use, and includes two issued US patents,
two pending US non-provisional patent applications, and twenty-six
pending foreign patent applications. The second family covers
additional SHP2 inhibitor compositions, their preparation and
methods of use, and includes one pending US non-provisional
application and six pending foreign patent applications. The
granted patent and any further patents that issue from these
applications from the two families are expected to expire in 2039,
absent any patent term adjustments or extensions.
As of December 31, 2021, we also own seven patent families relating
to ERAS-601. These families include ten pending US provisional
patent applications and three pending PCT applications. Any patents
issued from these applications are expected to expire between 2041
and 2042, absent any patent term adjustments or
extensions.
ERAS-3490
As of December 31, 2021, we own six patent families relating to
KRAS G12C inhibitors, their preparation, and method of use. These
patent families include four pending US non-provisional patent
applications, eight pending US provisional patent applications,
five pending PCT applications, and two pending foreign
applications. Any patents issued from these applications are
expected to expire between 2040 and 2042, absent any patent term
adjustments or extensions.
59
ERAS-801
As of December 31, 2021, we have sub-licensed three patent families
from Katmai, which Katmai in-licensed from the University of
California, Los Angeles (UCLA). Two of the patent families relate
to EGFR inhibitor compositions, their preparation, and methods of
use. The first patent family includes one pending US
non-provisional patent application, and eight pending foreign
patent applications. The second patent family covers the ERAS-801
product candidate and includes one pending US non-provisional
application and twenty-three pending foreign patent applications.
The third patent family relates to a functional companion assay for
brain cancer therapy and includes one pending PCT application. Any
patents issued from the first family of applications are expected
to expire in 2038, any patents issued from the second family of
applications are expected to expire in 2040, and any patents issued
from the third family of applications are expected to expire in
2041, in each case, absent any patent term adjustments or
extensions.
As of December 31, 2021, we also co-own with UCLA one patent family
relating to EGFR inhibitor compositions, their preparation and
methods of use. This patent family includes one pending PCT
application. Any patents issued from this application are expected
to expire in 2041, absent any patent term adjustments or
extensions.
As of December 31, 2021, we also own one patent family relating to
EGFR inhibitor polymorph forms. This patent family includes one
pending US provisional patent application. Any patents issued from
this application are expected to expire in 2042, absent any patent
term adjustments or extensions.
ERAS-2/3
As of December 31, 2021, we have in-licensed one patent family from
UCSF relating to covalent inhibitors of GTP- and GDP-bound RAS,
their preparation, and method of use. This patent family includes
one issued US patent, one pending US non-provisional patent
application and 13 pending foreign patent applications. The granted
patent and any further patents that issue from these applications
are expected to expire in 2037, absent any patent term adjustments
or extensions.
ERAS-4
As of December 31, 2021, we own three patent families relating to
KRAS G12D inhibitors, their preparation, and method of use. These
patent families include six pending US provisional patent
applications. Any patents issued from these applications are
expected to expire in 2042, absent any patent term adjustments or
extensions.
ERAS-5
As of December 31, 2021, we have in-licensed one patent family from
LifeArc relating to ULK1/2 inhibitors, their preparation, and
method of use. The patent family includes one pending US
provisional patent application. Any patents issued from this
application are expected to expire in 2042, absent any patent term
adjustments or extensions.
As of December 31, 2021, we also own one patent family relating to
ULK1/2 inhibitors, their preparation and method of use. This family
includes one pending US provisional patent application. Any patents
that issue from this application are expected to expire in 2042,
absent any patent term adjustments or extensions.
ERAS-10
As of December 31, 2021, we own two patent families relating to
PROTAC conjugates with undisclosed RAS/MAPK pathway target(s),
their preparation, and method of use. These patent families
includes seven pending US provisional patent applications. Any
patents issued from these applications are expected to expire in
2042, absent any patent term adjustments or extensions.
ERAS-12
As of December 31, 2021, we own one patent family relating to EGFR
D2/D3 bispecific antibodies, their preparation, and method of use.
The patent family includes one pending US provisional patent
application. Any patents issued from this application are expected
to expire in 2042, absent any patent term adjustments or
extensions.
60
Other IP programs or patents
With respect to our product candidates and processes we intend to
develop and commercialize in the normal course of business, we
intend to pursue patent protection covering, when possible,
compositions, methods of use, dosing and formulations. We may also
pursue patent protection with respect to manufacturing and drug
development processes and technologies. Obtaining and maintaining
patent protection depends on compliance with various procedural,
document submission, fee payment, and other requirements imposed by
governmental patent agencies. We may not be able to obtain patent
protections for our compositions, methods of use, dosing and
formulations, manufacturing and drug development processes and
technologies throughout the world. Issued patents can provide
protection for varying periods of time, depending upon the date of
filing of the patent application, the date of patent issuance and
the legal term of patents in the countries in which they are
obtained. In general, patents issued for applications filed in the
United States can provide exclusionary rights for 20 years from the
earliest effective filing date. In addition, in certain instances,
the term of an issued US patent that covers or claims an
FDA-approved product can be extended to recapture a portion of the
term effectively lost as a result of the FDA regulatory review
period, which is called patent term extension. The restoration
period cannot be longer than five years and the total patent term,
including the restoration period, must not exceed 14 years
following FDA approval. The US Patent and Trademark Office (USPTO)
may also adjust the term of a US patent to accommodate for delays
caused by the USPTO during the prosecution of a US patent
application. Congress has defined the conditions upon which an
applicant can receive an adjustment to the term and such
requirements are established in 35 USC 154(b). Similar provisions
are available in Europe and other jurisdictions to extend the term
of a patent that covers an approved drug. The term of patents
outside of the United States varies in accordance with the laws of
the foreign jurisdiction, but typically is also 20 years from the
earliest effective filing date. However, the actual protection
afforded by a patent varies on a product-by-product basis, from
country-to-country, and depends upon many factors, including the
type of patent, the scope of its coverage, the availability of
regulatory-related extensions, the availability of legal remedies
in a particular country, and the validity and enforceability of the
patent. Patent terms may be inadequate to protect our competitive
position on our products for an adequate amount of time. In the
future, if and when our therapeutic candidates receive FDA
approval, we expect to apply for patent term extensions on patents
covering those therapeutic candidates. We intend to seek patent
term extensions in any jurisdiction where these are available and
where we also have a patent that may be eligible; however, there is
no guarantee that the applicable authorities, including the USPTO
and FDA, will agree with our assessment of whether such extensions
should be granted, and even if granted, the length of such
extensions.
The patent positions of companies like ours are generally uncertain
and involve complex legal and factual questions. No consistent
policy regarding the scope of claims allowable in patents in the
field of biopharmaceuticals has emerged in the United States. The
relevant patent laws and their interpretation outside of the United
States is also uncertain. Changes in either the patent laws or
their interpretation in the United States and other countries may
diminish our ability to protect our technology or product
candidates and could affect the value of such intellectual
property. In particular, our ability to stop third parties from
making, using, selling, offering to sell or importing products that
infringe our intellectual property will depend in part on our
success in obtaining and enforcing patent claims that cover our
technology, inventions and improvements. We cannot guarantee that
patents will be granted with respect to any of our pending patent
applications or with respect to any patent applications we may file
in the future, nor can we be sure that any patents that may be
granted to us in the future will be commercially useful in
protecting our products, the methods of use or manufacture of those
products. Moreover, even our issued patents do not guarantee us the
right to practice our technology in relation to the
commercialization of our products. Patent and other intellectual
property rights in the pharmaceutical and biotechnology space are
evolving and involve many risks and uncertainties. For example,
third parties may have blocking patents that could be used to
prevent us from commercializing our product candidates and
practicing our proprietary technology, and our issued patents may
be challenged, invalidated or circumvented, which could limit our
ability to stop competitors from marketing related products or
could limit the term of patent protection that otherwise may exist
for our product candidates. In addition, the scope of the rights
granted under any issued patents may not provide us with protection
or competitive advantages against competitors with similar
technology. Furthermore, our competitors may independently develop
similar technologies that are outside the scope of the rights
granted under any issued patents. For these reasons, we may face
competition with respect to our product candidates. Moreover,
because of the extensive time required for development, testing and
regulatory review of a potential product, it is possible that,
before any particular product candidate can be commercialized, any
patent protection for such product may expire or remain in force
for only a short period following commercialization, thereby
reducing the commercial advantage the patent provides.
We also rely on trade secrets to protect aspects of our technology
and business not amenable to, or that we do not consider
appropriate for, patent protection. We seek to protect this
intellectual property, in part, by requiring our employees,
consultants, outside scientific collaborators, sponsored
researchers and other service providers and advisors to execute
confidentiality agreements upon the commencement of employment or
other relationship with us. In general, these agreements provide
that confidential information concerning our business or financial
affairs developed or made
61
known to the individual during the course of the individual’s
relationship with us is to be kept confidential and not disclosed
to third parties except in specific circumstances. In the case of
employees, the agreements further provide that inventions and
discoveries conceived or reduced to practice by the individual that
are related to our business, or actual, or demonstrably
anticipated, research or development, or made during normal working
hours, on our premises or using our equipment, supplies, or
proprietary information, are our exclusive property. In many cases
our agreements with consultants, outside scientific collaborators,
sponsored researchers and other service providers and advisors
require them to assign, or grant us licenses to, inventions
resulting from the work or services they render under such
agreements or grant us an option to negotiate a license to use such
inventions.
We seek trademark protection in the United States and in certain
other jurisdictions where available and when we deem appropriate.
We currently have registrations for our “ERASCA” mark in the United
States as well as in over 20 foreign jurisdictions, including the
European Union. We have also filed a trademark application in the
United States as well as over 20 foreign jurisdictions for
registration of our “MAPKLAMP” mark.
Manufacturing
We do not own or operate, and currently have no plans to establish,
any manufacturing facilities. We rely, and expect to continue to
rely, on third parties for the manufacture of our product
candidates for preclinical and clinical testing, as well as for
commercial manufacture if any of our product candidates obtain
marketing approval. We are working with our current manufacturers
to ensure that we will be able to scale up our manufacturing
capabilities to support our clinical plans. We are also in the
process of locating and qualifying additional manufacturers to
build redundancies into our supply chain. In addition, we rely on
third parties to package, label, store, and distribute our product
candidates, and we intend to rely on third parties for our
commercial products if marketing approval is obtained. We believe
that this strategy allows us to maintain a more efficient
infrastructure by eliminating the need for us to invest in our own
manufacturing facilities, equipment, and personnel while also
enabling us to focus our expertise and resources on the design and
development of our product candidates.
Government Regulation
The FDA and comparable regulatory authorities in state and local
jurisdictions and in other countries impose substantial and
burdensome requirements upon companies involved in the clinical
development, manufacture, marketing and distribution of drugs and
biologics such as those we are developing. These entities regulate,
among other things, the research and development, testing,
manufacture, quality control, safety, effectiveness, labeling,
storage, record keeping, approval, advertising and promotion,
distribution, post-approval monitoring and reporting, sampling and
export and import of our product candidates.
US regulation of drugs and biologics
In the United States, the FDA regulates drugs under the Federal
Food, Drug, and Cosmetic Act (FDCA), and its implementing
regulations, and biologics under the FDCA and the Public Health
Service Act and their implementing regulations. FDA approval of a
new drug application (NDA) or biologics license application (BLA)
or supplement is required before any new unapproved drug, biologic
or dosage form, including a new use of a previously approved drug
or biologic, can be marketed in the United States.
The process required by the FDA before such product candidates may
be marketed in the United States generally involves the
following:
•
completion of extensive preclinical laboratory tests and
preclinical animal studies, performed in accordance with applicable
Good Laboratory Practice (GLP) regulations;
•
submission to the FDA of an investigational new drug application
(IND) which must become effective before human clinical studies may
begin and must be updated annually;
•
approval by an independent institutional review board (IRB) or
ethics committee representing each clinical site before each
clinical study may be initiated;
•
performance of adequate and well-controlled human clinical studies
in accordance with Good Clinical Practice (GCP) requirements to
establish the safety and efficacy, or with respect to biologics,
the safety, purity and potency of the product candidate for each
proposed indication;
•
preparation of and submission to the FDA of an NDA or BLA, after
completion of all pivotal clinical studies;
62
•
potential review of the product application by an FDA advisory
committee, where appropriate and if applicable;
•
a determination by the FDA within 60 days of its receipt of an NDA
or BLA to file the application for review;
•
satisfactory completion of an FDA pre-approval inspection of the
manufacturing facilities where the proposed product drug substance
is produced to assess compliance with current Good Manufacturing
Practice requirements (cGMPs) and audits of selected clinical trial
sites to ensure compliance with GCP; and
•
FDA review and approval of an NDA or BLA prior to any commercial
marketing or sale of the drug in the United States.
Preclinical studies include laboratory evaluation of product
chemistry, toxicity and formulation, as well as
in vitro
and animal studies to assess potential safety and efficacy. The
conduct of preclinical studies is subject to federal regulations
and requirements, including GLP regulations applicable to certain
safety/toxicology studies.
An IND is a request for authorization from the FDA to administer an
investigational new drug product to humans. The central focus of an
IND submission is on the general investigational plan and the
protocol or protocols for preclinical studies and clinical trials.
The IND also includes results of animal and in vitro studies
assessing the toxicology, pharmacokinetics, pharmacology and
pharmacodynamic characteristics of the product, chemistry,
manufacturing and controls (CMC) information, and any available
human data or literature to support the use of the investigational
product. An IND must become effective before human clinical trials
may begin. The IND automatically becomes effective 30 days after
receipt by the FDA, unless the FDA, within the 30-day period,
raises safety concerns or questions about the proposed clinical
trial. In such a case, the IND may be placed on clinical hold and
the IND sponsor and the FDA must resolve any outstanding concerns
or questions before the clinical trial can begin. Submission of an
IND therefore may or may not result in FDA authorization to begin a
clinical trial.
Clinical trials involve the administration of the investigational
product to human subjects under the supervision of qualified
investigators in accordance with GCP, which includes the
requirement that all research subjects, or their legal
representative, provide their informed consent for their
participation in any clinical study. Clinical trials are conducted
under protocols detailing, among other things, the inclusion and
exclusion criteria, the objectives of the study, the parameters to
be used in monitoring safety and the effectiveness criteria to be
evaluated. A separate submission to the existing IND must be made
for each successive clinical trial conducted during product
development and for any subsequent protocol amendments.
Furthermore, an independent IRB for each site proposing to conduct
the clinical trial must review and approve the plan for any
clinical trial and its informed consent form before the clinical
trial begins at that site, and must monitor the study until
completed. An IRB is charged with protecting the welfare and rights
of trial participants and considers such items as whether the risks
to individuals participating in the clinical trials are minimized
and are reasonable in relation to anticipated benefits. Regulatory
authorities, the IRB or the sponsor may suspend a clinical trial at
any time on various grounds, including a finding that the subjects
are being exposed to an unacceptable health risk or that the trial
is unlikely to meet its stated objectives. Some studies also
include oversight by an independent group of qualified experts
organized by the clinical study sponsor, known as a data safety
monitoring board or committee, which provides authorization for
whether or not a study may move forward at designated check points
based on access to certain data from the study and may recommend
that the clinical trial be halted if it determines that there is an
unacceptable safety risk for subjects or other grounds, such as no
demonstration of efficacy. There are also requirements governing
the reporting of clinical trials and clinical study results to
public registries.
The clinical investigation of a drug is generally divided into
three phases. Although the phases are usually conducted
sequentially, they may overlap or be combined.
•
Phase 1:
The investigational product is initially introduced into healthy
human subjects or patients with the target disease or
condition.
These studies are designed to test the safety, dosage tolerance,
absorption, metabolism and distribution of the investigational
product in humans, the side effects associated with increasing
doses, and, if possible, to gain early evidence on
effectiveness.
•
Phase 2:
The investigational product is administered to a limited patient
population with a specified disease or condition to evaluate
the
preliminary efficacy, optimal dosages and dosing schedule and to
identify possible adverse side effects and safety risks. Multiple
Phase 2 clinical trials may be conducted to obtain information
prior to beginning larger and more expensive Phase 3 clinical
trials.
63
•
Phase 3:
The investigational product is administered to an expanded patient
population to further evaluate dosage, to provide
statistically
significant evidence of clinical efficacy and to further test for
safety, generally at multiple geographically dispersed clinical
trial sites. These clinical trials are intended to establish the
overall risk/benefit ratio of the investigational product and to
provide an adequate basis for labeling.
Post-marketing studies, sometimes referred to as Phase 4 clinical
trials, may be conducted after initial marketing approval. These
clinical trials are used to gain additional experience from the
treatment of patients in the intended therapeutic indication. In
certain instances, such as with accelerated approval drugs, the FDA
may mandate the performance of Phase 4 clinical trials as a
condition of approval.
Concurrent with clinical trials, companies may complete additional
animal studies and develop additional information about the
biological characteristics of the product candidate, and must
finalize a process for manufacturing the product in commercial
quantities in accordance with cGMP requirements. The manufacturing
process must be capable of consistently producing quality batches
of the product candidate and, among other things, must develop
methods for testing the identity, strength, quality and purity of
the final product, or for biologics, the safety, purity and
potency.
While the IND is active, progress reports summarizing the results
of the clinical trials and nonclinical studies performed since the
last progress report, among other things, must be submitted at
least annually to the FDA, and written IND safety reports must be
submitted to the FDA and investigators for serious and unexpected
suspected adverse events, findings from other studies suggesting a
significant risk to humans exposed to the same or similar drugs or
biologics, findings from animal or
in vitro
testing suggesting a significant risk to humans, and any clinically
important increased incidence of a serious suspected adverse
reaction compared to that listed in the protocol or investigator
brochure.
In addition, during the development of a new drug or biologic,
sponsors are given opportunities to meet with the FDA at certain
points. These points may be prior to submission of an IND, at the
end of Phase 2, and before an NDA or BLA is submitted. Meetings at
other times may be requested. These meetings can provide an
opportunity for the sponsor to share information about the data
gathered to date, for the FDA to provide advice, and for the
sponsor and the FDA to reach agreement on the next phase of
development. Sponsors typically use the meetings at the end of the
Phase 2 trial to discuss Phase 2 clinical results and present plans
for the pivotal Phase 3 clinical trials that they believe will
support approval of the new drug or biologic.
NDA and BLA review process
Assuming successful completion of all required testing in
accordance with all applicable regulatory requirements, the results
of product development, nonclinical studies and clinical trials are
submitted to the FDA as part of an NDA or BLA requesting approval
to market the product for one or more indications. The submission
of an NDA or BLA requires payment of a substantial application user
fee to the FDA, unless a waiver or exemption applies.
The NDA or BLA must include all relevant data available from
pertinent preclinical studies and clinical trials, including
negative or ambiguous results as well as positive findings,
together with detailed information relating to the product’s CMC
and proposed labeling, among other things. Data can come from
company-sponsored clinical studies intended to test the safety and
effectiveness of the product, or from a number of alternative
sources, including studies initiated and sponsored by
investigators.
In addition, under the Pediatric Research Equity Act (PREA), an NDA
or BLA or supplement to an NDA or BLA must contain data to assess
the safety and effectiveness of the biological product candidate
for the claimed indications in all relevant pediatric
subpopulations and to support dosing and administration for each
pediatric subpopulation for which the product is safe and
effective. The Food and Drug Administration Safety and Innovation
Act requires that a sponsor who is planning to submit a marketing
application for a drug or biological product that includes a new
active ingredient, new indication, new dosage form, new dosing
regimen or new route of administration submit an initial pediatric
study plan within sixty days after an end-of-Phase 2 meeting or as
may be agreed between the sponsor and FDA. Unless otherwise
required by regulation, PREA does not apply to any drug or
biological product for an indication for which orphan designation
has been granted.
Within 60 days following submission of the application, the FDA
reviews the submitted BLA or NDA to determine if the application is
substantially complete before the agency accepts it for filing. The
FDA may refuse to file any NDA or BLA that it deems incomplete or
not properly reviewable at the time of submission and may request
additional information. In this event, the NDA or BLA must be
resubmitted with the additional information. Once an NDA or BLA has
been accepted for filing, the FDA’s goal is to review standard
applications within ten months after the filing date, or, if the
application qualifies for priority review, six months after the FDA
accepts the application for filing. In both standard and
priority
64
reviews, the review process may also be extended by FDA requests
for additional information or clarification. Once accepted for
filing, the FDA reviews an NDA to determine, among other things,
whether a product is safe and effective for its intended use and
whether its manufacturing is sufficient to assure and preserve the
product’s identity, strength, quality and purity. The FDA reviews a
BLA to determine, among other things, whether a product is safe,
pure and potent and the facility in which it is manufactured,
processed, packed or held meets standards designed to assure the
product’s continued safety, purity and potency. When reviewing an
NDA or BLA, the FDA may convene an advisory committee to provide
clinical insight on application review questions. The FDA is not
bound by the recommendations of an advisory committee, but it
considers such recommendations carefully when making
decisions.
Before approving an NDA or BLA, the FDA will typically inspect the
facility or facilities where the product is manufactured. The FDA
will not approve an application unless it determines that the
manufacturing processes and facilities are in compliance with cGMP
requirements and adequate to assure consistent production of the
product within required specifications. Additionally, before
approving an NDA or BLA, the FDA will typically inspect one or more
clinical sites to assure compliance with GCP.
After the FDA evaluates the NDA or BLA and conducts inspections of
manufacturing facilities where the investigational product and/or
its drug substance will be produced, the FDA may issue an approval
letter or a Complete Response Letter (CRL). An approval letter
authorizes commercial marketing of the product with specific
prescribing information for specific indications. A CRL signals
that the review cycle is complete and the application cannot be
approved in its present form. The CRL will generally describe all
of the deficiencies that the FDA has identified in the NDA or BLA,
except that where the FDA determines that the data supporting the
application are inadequate to support approval, the FDA may issue
the CRL without first conducting required inspections, testing
submitted product lots and/or reviewing proposed labeling. In
issuing the CRL, the FDA may recommend actions that the applicant
might take to place the NDA or BLA in condition for approval,
including requests for additional information or clarification. The
FDA may delay or refuse approval of an NDA or BLA if applicable
regulatory criteria are not satisfied, require additional testing
or information and/or require post-marketing testing and
surveillance to monitor safety or efficacy of a product.
If regulatory approval of a product is granted, such approval will
be granted for particular indications and may entail limitations on
the indicated uses for which such product may be marketed. For
example, the FDA may approve the NDA or BLA with a Risk Evaluation
and Mitigation Strategy (REMS) to ensure the benefits of the
product outweigh its risks. A REMS is a safety strategy to manage a
known or potential serious risk associated with a product and to
enable patients to have continued access to such medicines by
managing their safe use, and could include medication guides,
physician communication plans, or elements to assure safe use, such
as restricted distribution methods, patient registries and other
risk minimization tools. The FDA also may condition approval on,
among other things, changes to proposed labeling or the development
of adequate controls and specifications. Once approved, the FDA may
withdraw the product approval if compliance with pre- and
post-marketing requirements is not maintained or if problems occur
after the product reaches the marketplace. The FDA may require one
or more Phase 4 post-market studies and surveillance to further
assess and monitor the product’s safety and effectiveness after
commercialization, and may limit further marketing of the product
based on the results of these post-marketing studies.
Expedited development and review programs
The FDA offers a number of expedited development and review
programs for qualifying product candidates. For example, the fast
track program is intended to expedite or facilitate the process for
reviewing product candidates that meet certain criteria.
Specifically, product candidates are eligible for fast track
designation if they are intended to treat a serious or
life-threatening disease or condition and demonstrate the potential
to address unmet medical needs for the disease or condition. Fast
track designation applies to the combination of the product
candidate and the specific indication for which it is being
studied. The sponsor of a fast track product candidate has
opportunities for more frequent interactions with the review team
during product development and, once an NDA or BLA is submitted,
the product may be eligible for priority review, if the relevant
criteria are met. A fast track product candidate may also be
eligible for rolling review, where the FDA may consider for review
sections of the NDA or BLA on a rolling basis before the complete
application is submitted, if the sponsor provides a schedule for
the submission of the sections of the NDA or BLA, the FDA agrees to
accept sections of the NDA or BLA and determines that the schedule
is acceptable, and the sponsor pays any required user fees upon
submission of the first section of the NDA or BLA.
A product candidate intended to treat a serious or life-threatening
disease or condition may also be eligible for breakthrough therapy
designation to expedite its development and review. A product can
receive breakthrough therapy designation if preliminary clinical
evidence indicates that the product candidate, alone or in
combination with one or more other drugs or biologics, may
demonstrate substantial improvement over existing therapies on one
or more clinically significant endpoints, such as substantial
treatment effects observed early in clinical development. The
designation
65
includes all of the fast track program features, as well as more
intensive FDA interaction and guidance beginning as early as Phase
1 and an organizational commitment to expedite the development and
review of the product candidate, including involvement of senior
managers.
Any marketing application for a drug or biologic submitted to the
FDA for approval, including a product candidate with a fast track
designation and/or breakthrough therapy designation, may be
eligible for other types of FDA programs intended to expedite the
FDA review and approval process, such as priority review and
accelerated approval. An NDA or BLA is eligible for priority review
if the product candidate has the potential to provide a significant
improvement in the treatment, diagnosis or prevention of a serious
disease or condition. For new molecular entity NDAs and original
BLAs, priority review designation means the FDA’s goal is to take
action on the marketing application within six months of the 60-day
filing date (as compared to 10 months under standard
review).
Additionally, product candidates studied for their safety and
effectiveness in treating serious or life-threatening diseases or
conditions may receive accelerated approval upon a determination
that the product candidate has an effect on a surrogate endpoint
that is reasonably likely to predict clinical benefit, or on a
clinical endpoint that can be measured earlier than irreversible
morbidity or mortality, that is reasonably likely to predict an
effect on irreversible morbidity or mortality or other clinical
benefit, taking into account the severity, rarity, or prevalence of
the condition and the availability or lack of alternative
treatments. As a condition of accelerated approval, the FDA will
generally require the sponsor to perform adequate and
well-controlled post-marketing clinical studies to verify and
describe the anticipated effect on irreversible morbidity or
mortality or other clinical benefit. Products receiving accelerated
approval may be subject to expedited withdrawal procedures if the
sponsor fails to conduct the required post-marketing studies or if
such studies fail to verify the predicted clinical benefit. In
addition, the FDA currently requires as a condition for accelerated
approval pre-approval of promotional materials, which could
adversely impact the timing of the commercial launch of the
product.
Fast track designation, breakthrough therapy designation, priority
review, and accelerated approval do not change the standards for
approval but may expedite the development or approval process. Even
if a product qualifies for one or more of these programs, the FDA
may later decide that the product no longer meets the conditions
for qualification or decide that the time period for FDA review or
approval will not be shortened.
Orphan drug designation
Under the Orphan Drug Act, the FDA may grant orphan designation to
a drug or biologic intended to treat a rare disease or condition,
which is a disease or condition that affects fewer than 200,000
individuals in the United States, or more than 200,000 individuals
in the United States for which there is no reasonable expectation
that the cost of developing and making available in the United
States a drug or biologic for this type of disease or condition
will be recovered from sales in the United States for that drug or
biologic. Orphan drug designation must be requested before
submitting an NDA or BLA. After the FDA grants orphan drug
designation, the generic identity of the therapeutic agent and its
potential orphan use are disclosed publicly by the FDA. The orphan
drug designation does not convey any advantage in, or shorten the
duration of, the regulatory review or approval process.
If a product candidate that has orphan drug designation
subsequently receives the first FDA approval for the disease for
which it has such designation, the product is entitled to orphan
drug exclusive approval (or exclusivity), which means that the FDA
may not approve any other applications, including a full NDA or
BLA, to market the same drug or biologic for the same indication
for seven years, except in limited circumstances, such as a showing
of clinical superiority to the product with orphan drug exclusivity
or if the FDA finds that the holder of the orphan drug exclusivity
has not shown that it can assure the availability of sufficient
quantities of the orphan drug to meet the needs of patients with
the disease or condition for which the drug or biologic was
designated. Orphan drug exclusivity does not prevent the FDA from
approving a different drug or biologic for the same disease or
condition, or the same drug or biologic for a different disease or
condition. Among the other benefits of orphan drug designation are
tax credits for certain research and a waiver of the NDA or BLA
application user fee.
A designated orphan drug may not receive orphan drug exclusivity if
it is approved for a use that is broader than the indication for
which it received orphan designation. In addition, exclusive
marketing rights in the United States may be lost if the FDA later
determines that the request for designation was materially
defective or if the manufacturer is unable to assure sufficient
quantities of the product to meet the needs of patients with the
rare disease or condition.
66
Post-approval requirements
Any products manufactured or distributed pursuant to FDA approvals
are subject to pervasive and continuing regulation by the FDA,
including, among other things, requirements relating to
record-keeping, reporting of adverse experiences, periodic
reporting, product sampling and distribution, and advertising and
promotion of the product. After approval, most changes to the
approved product, such as adding new indications or other labeling
claims, are subject to prior FDA review and approval. There also
are continuing user fee requirements, under which the FDA assesses
an annual program fee for each product identified in an approved
NDA or BLA. Drug and biologic manufacturers and their
subcontractors are required to register their establishments with
the FDA and certain state agencies, and are subject to periodic
unannounced inspections by the FDA and certain state agencies for
compliance with cGMPs, which impose certain procedural and
documentation requirements upon us and our third-party
manufacturers. Changes to the manufacturing process are strictly
regulated, and, depending on the significance of the change, may
require prior FDA approval before being implemented. FDA
regulations also require investigation and correction of any
deviations from cGMPs and impose reporting requirements upon us and
any third-party manufacturers that we may decide to use.
Accordingly, manufacturers must continue to expend time, money and
effort in the area of production and quality control to maintain
compliance with cGMPs and other aspects of regulatory
compliance.
The FDA may withdraw approval if compliance with regulatory
requirements and standards is not maintained or if problems occur
after the product reaches the market. Later discovery of previously
unknown problems with a product, including adverse events of
unanticipated severity or frequency, or with manufacturing
processes, or failure to comply with regulatory requirements, may
result in revisions to the approved labeling to add new safety
information; imposition of post-market studies or clinical studies
to assess new safety risks; or imposition of distribution
restrictions or other restrictions under a REMS program. Other
potential consequences include, among other things:
•
restrictions on the marketing or manufacturing of a product,
complete withdrawal of the product from the market or product
recalls;
•
fines, warning letters or holds on post-approval clinical
studies;
•
refusal of the FDA to approve pending applications or supplements
to approved applications, or suspension or revocation of existing
product approvals;
•
product seizure or detention, or refusal of the FDA to permit the
import or export of products;
•
consent decrees, corporate integrity agreements, debarment or
exclusion from federal healthcare programs;
•
mandated modification of promotional materials and labeling and the
issuance of corrective information;
•
injunctions or the imposition of civil or criminal
penalties.
The FDA closely regulates the marketing, labeling, advertising and
promotion of drug products. A company can make only those claims
relating to safety and efficacy, purity and potency that are
approved by the FDA and in accordance with the provisions of the
approved label. The FDA and other agencies actively enforce the
laws and regulations prohibiting the promotion of off-label uses.
Failure to comply with these requirements can result in, among
other things, adverse publicity, warning letters, corrective
advertising and potential civil and criminal penalties. Physicians
may prescribe, in their independent professional medical judgment,
legally available products for uses that are not described in the
product’s labeling and that differ from those tested by us and
approved by the FDA. Physicians may believe that such off-label
uses are the best treatment for many patients in varied
circumstances. The FDA does not regulate the behavior of physicians
in their choice of treatments. The FDA does, however, restrict
manufacturer’s communications on the subject of off-label use of
their products. The federal government has levied large civil and
criminal fines against companies for alleged improper promotion of
off-label use and has enjoined companies from engaging in off-label
promotion. The FDA and other regulatory agencies have also required
that companies enter into consent decrees or permanent injunctions
under which specified promotional conduct is changed or curtailed.
However, companies may share truthful and not misleading
information that is otherwise consistent with a product’s
FDA-approved labeling.
In addition, the distribution of prescription biopharmaceutical
products is subject to the Prescription Drug Marketing Act (PDMA)
which regulates the distribution of drugs and drug samples at the
federal level, and sets minimum standards for the registration and
regulation of drug distributors by the states. Both the PDMA and
state laws limit the distribution of prescription pharmaceutical
product samples and impose requirements to ensure accountability in
distribution.
67
Drug product marketing exclusivity
Market exclusivity provisions under the FDCA can delay the
submission or the approval of certain marketing applications. For
example, the FDCA provides a five-year period of non-patent
marketing exclusivity within the United States to the first
applicant to obtain approval of an NDA for a new chemical entity. A
drug is a new chemical entity if the FDA has not previously
approved any other new drug containing the same active moiety,
which is the molecule or ion responsible for the action of the drug
substance. During the exclusivity period, the FDA may not approve
or even accept for review an abbreviated new drug application
(ANDA), or an NDA submitted under Section 505(b)(2), or 505(b) (2)
NDA, submitted by another company for another drug based on the
same active moiety, regardless of whether the drug is intended for
the same indication as the original innovative drug or for another
indication, where the applicant does not own or have a legal right
of reference to all the data required for approval. However, an
application may be submitted after four years if it contains a
certification of patent invalidity or non-infringement to one of
the patents listed with the FDA by the innovator NDA
holder.
The FDCA alternatively provides three years of marketing
exclusivity for an NDA, or supplement to an existing NDA if new
clinical investigations, other than bioavailability studies, that
were conducted or sponsored by the applicant are deemed by the FDA
to be essential to the approval of the application, for example new
indications, dosages or strengths of an existing drug. This
three-year exclusivity covers only the modification for which the
drug received approval on the basis of the new clinical
investigations and does not prohibit the FDA from approving ANDAs
or 505(b)(2) NDAs for drugs containing the active agent for the
original indication or condition of use. Five-year and three-year
exclusivity will not delay the submission or approval of a full
NDA. However, an applicant submitting a full NDA would be required
to conduct or obtain a right of reference to any preclinical
studies and adequate and well-controlled clinical trials necessary
to demonstrate safety and effectiveness.
Pediatric exclusivity is another type of marketing exclusivity
available in the United States. Pediatric exclusivity provides for
an additional six months of marketing exclusivity attached to
another period of exclusivity if a sponsor conducts clinical trials
in children in response to a Written Request from the FDA. The
issuance of a written request does not require the sponsor to
undertake the described clinical trials. In addition, orphan drug
exclusivity, as described above, may offer a seven-year period of
marketing exclusivity, except in certain circumstances.
Biosimilars and reference product exclusivity
The Biologics Price Competition and Innovation Act of 2009 (BPCIA)
created an abbreviated approval pathway for biological products
that are highly similar, or “biosimilar,” to or interchangeable
with an FDA-approved reference biological product. Biosimilarity,
which requires that there be no clinically meaningful differences
between the biological product and the reference product in terms
of safety, purity, and potency, is generally shown through
analytical studies, animal studies, and a clinical study or
studies. Interchangeability requires that a product is biosimilar
to the reference product and the product must demonstrate that it
can be expected to produce the same clinical results as the
reference product in any given patient and, for products that are
administered multiple times to an individual, the biologic and the
reference biologic may be alternated or switched after one has been
previously administered without increasing safety risks or risks of
diminished efficacy relative to exclusive use of the reference
biologic. A product shown to be biosimilar or interchangeable with
an FDA-approved reference biological product may rely in part on
the FDA’s previous determination of safety and effectiveness for
the reference product for approval, which can potentially reduce
the cost and time required to obtain approval to market the
product.
Under the BPCIA, an application for a biosimilar product may not be
submitted to the FDA until four years following the date that the
reference product was first licensed by the FDA. In addition, the
approval of a biosimilar product may not be made effective by the
FDA until 12 years from the date on which the reference product was
first licensed. During this 12-year period of exclusivity, another
company may still market a competing version of the reference
product if the FDA approves a full BLA for the competing product
containing that applicant’s own preclinical data and data from
adequate and well-controlled clinical trials to demonstrate the
safety, purity and potency of its product. The BPCIA also created
certain exclusivity periods for biosimilars approved as
interchangeable products. At this juncture, it is unclear whether
products deemed “interchangeable” by the FDA will, in fact, be
readily substituted by pharmacies, which are governed by state
pharmacy law.
A biological product can also obtain pediatric market exclusivity
in the United States. Pediatric exclusivity, if granted, adds six
months to existing exclusivity periods and patent terms. This
six-month exclusivity, which runs from the end of other exclusivity
protection or patent term, may be granted based on the voluntary
completion of a pediatric study in accordance with an FDA-issued
“Written Request” for such a study.
68
FDA regulation of companion diagnostics
If safe and effective use of a drug or biologic depends on
an
in vitro
diagnostic, then the FDA may require approval or clearance of that
diagnostic, known as a companion diagnostic, at the same time that
the FDA approves the therapeutic product. In August 2014, the FDA
issued final guidance clarifying the requirements that will apply
to approval of therapeutic products and in vitro companion
diagnostics. According to the guidance, if FDA determines that a
companion diagnostic device is essential to the safe and effective
use of a novel therapeutic product or indication, FDA may will not
approve the drug or new indication if the companion diagnostic
device is not also approved or cleared for that indication.
Approval or clearance of the companion diagnostic device will
ensure that the device has been adequately evaluated and has
adequate performance characteristics in the intended population.
The review of
in vitro
companion diagnostics in conjunction with the review of our product
candidates will, therefore, likely involve coordination of review
by the FDA’s Center for Drug Evaluation and Research or the FDA’s
Center for Biologics Evaluation and Research and the FDA’s Center
for Devices and Radiological Health Office of In Vitro Diagnostics
and Radiological Health.
Under the FDCA,
in vitro
diagnostics, including companion diagnostics, are regulated as
medical devices. In the United States, the FDCA and its
implementing regulations, and other federal and state statutes and
regulations govern, among other things, medical device design and
development, preclinical and clinical testing, premarket clearance
or approval, registration and listing, manufacturing, labeling,
storage, advertising and promotion, sales and distribution, export
and import, and post-market surveillance. Unless an exemption
applies, diagnostic tests require marketing clearance or approval
from the FDA prior to commercial distribution. The two primary
types of FDA marketing authorization applicable to a medical device
are clearance of a premarket notification pursuant to Section
510(k) of the FDCA, also called 510(k) clearance, and approval of a
premarket approval application (PMA).
The PMA process, including the gathering of clinical and
preclinical data and the submission to and review by the FDA, can
take several years or longer. It involves a rigorous premarket
review during which the applicant must prepare and provide the FDA
with reasonable assurance of the device’s safety and effectiveness
and information about the device and its components regarding,
among other things, device design, manufacturing and labeling. PMA
applications are subject to an application fee. In addition, PMAs
for certain devices must generally include the results from
extensive preclinical and adequate and well-controlled clinical
trials to establish the safety and effectiveness of the device for
each indication for which FDA approval is sought. As part of the
PMA review, the FDA will typically inspect the manufacturer’s
facilities for compliance with the Quality System Regulation (QSR)
which imposes elaborate testing, control, documentation and other
quality assurance requirements.
If the FDA’s evaluation of the PMA application is favorable, the
FDA typically issues an approvable letter requiring the applicant’s
agreement to specific conditions, such as changes in labeling, or
specific additional information, such as submission of final
labeling, in order to secure final approval of the PMA. If the
FDA’s evaluation of the PMA or manufacturing facilities is not
favorable, the FDA will deny approval of the PMA or issue a not
approvable letter. A not approvable letter will outline the
deficiencies in the application and, where practical, will identify
what is necessary to make the PMA approvable. The FDA may also
determine that additional clinical trials are necessary, in which
case the PMA approval may be delayed for several months or years
while the trials are conducted and then the data submitted in an
amendment to the PMA. If the FDA concludes that the applicable
criteria have been met, the FDA will issue a PMA for the approved
indications, which can be more limited than those originally sought
by the applicant. The PMA can include post-approval conditions that
the FDA believes necessary to ensure the safety and effectiveness
of the device, including, among other things, restrictions on
labeling, promotion, sale and distribution. Once granted, PMA
approval may be withdrawn by the FDA if compliance with
post-approval requirements, conditions of approval or other
regulatory standards are not maintained or problems are identified
following initial marketing.
After a device is placed on the market, it remains subject to
significant regulatory requirements. Medical devices may be
marketed only for the uses and indications for which they are
cleared or approved. Device manufacturers must also establish
registration and device listings with the FDA. A medical device
manufacturer’s manufacturing processes and those of its suppliers
are required to comply with the applicable portions of the QSR,
which cover the methods and documentation of the design, testing,
production, processes, controls, quality assurance, labeling,
packaging and shipping of medical devices. Domestic facility
records and manufacturing processes are subject to periodic
unscheduled inspections by the FDA. The FDA also may inspect
foreign facilities that export products to the United
States.
69
Other US regulatory requirements
In addition to FDA regulation of pharmaceutical products,
pharmaceutical companies are also subject to additional healthcare
regulation and enforcement by the federal government and by
authorities in the states and foreign jurisdictions in which they
conduct their business and may constrain the financial arrangements
and relationships through which we research, as well as sell,
market and distribute any products for which we obtain marketing
authorization. Such laws include, without limitation, state and
federal anti-kickback, fraud and abuse, false claims, data privacy
and security, and transparency laws and regulations related to drug
pricing and payments and other transfers of value made to
physicians and other healthcare providers. If their operations are
found to be in violation of any of such laws or any other
governmental regulations that apply, they may be subject to
penalties, including, without limitation, administrative, civil and
criminal penalties, damages, fines, disgorgement, the curtailment
or restructuring of operations, integrity oversight and reporting
obligations, exclusion from participation in federal and state
healthcare programs and imprisonment.
US coverage and reimbursement
Significant uncertainty exists as to the coverage and reimbursement
status of any product candidate for which we may seek regulatory
approval. Sales in the United States will depend, in part, on the
availability of sufficient coverage and adequate reimbursement from
third-party payors, which include government health programs such
as Medicare, Medicaid, TRICARE and the Veterans Administration, as
well as managed care organizations and private health insurers.
Prices at which we or our customers seek reimbursement for our
product candidates can be subject to challenge, reduction or denial
by third-party payors.
The process for determining whether a third-party payor will
provide coverage for a product is typically separate from the
process for setting the reimbursement rate that the payor will pay
for the product. In the United States, there is no uniform policy
among payors for coverage or reimbursement. Decisions regarding
whether to cover any of a product, the extent of coverage and
amount of reimbursement to be provided are made on a plan-by-plan
basis. Third-party payors often rely upon Medicare coverage policy
and payment limitations in setting their own coverage and
reimbursement policies, but also have their own methods and
approval processes. Therefore, coverage and reimbursement for
products can differ significantly from payor to payor. As a result,
the coverage determination process is often a time-consuming and
costly process that can require manufacturers to provide scientific
and clinical support for the use of a product to each payor
separately, with no assurance that coverage and adequate
reimbursement will be applied consistently or obtained in the first
instance. In addition, companion diagnostic tests require coverage
and reimbursement separate and apart from the coverage and
reimbursement for their companion pharmaceutical or biological
products. Similar challenges to obtaining coverage and
reimbursement, applicable to pharmaceutical or biological products,
will apply to companion diagnostics.
Third-party payors are increasingly challenging the price and
examining the medical necessity and cost-effectiveness of medical
products and services, in addition to their safety and efficacy.
Third-party payors may not consider our product candidates to be
medically necessary or cost-effective compared to other available
therapies. Adoption of price controls and cost-containment
measures, and adoption of more restrictive policies in
jurisdictions with existing controls and measures, could further
limit sales of any product that receives approval.
US healthcare reform
In the United States, there have been, and continue to be,
legislative and regulatory changes and proposed changes regarding
the healthcare system that could prevent or delay marketing
approval of product candidates, restrict or regulate post-approval
activities, and affect the profitable sale of product candidates.
Among policy makers and payors in the United States, there is
significant interest in promoting changes in healthcare systems
with the stated goals of containing healthcare costs, improving
quality and/or expanding access. In the United States, the
pharmaceutical industry has been a particular focus of these
efforts and has been significantly affected by major legislative
initiatives.
By way of example, in March 2010, the Patient Protection and
Affordable Care Act (the ACA) was passed, which substantially
changed the way healthcare is financed by both governmental and
private insurers, and significantly affected the pharmaceutical
industry. The ACA, among other things, increased the minimum level
of Medicaid rebates payable by manufacturers of brand name drugs
from 15.1% to 23.1% of the average manufacturer price; required
collection of rebates for drugs paid by Medicaid managed care
organizations; required manufacturers to participate in a coverage
gap discount program, in which manufacturers must agree to offer
point-of-sale discounts off negotiated prices of applicable brand
drugs to eligible beneficiaries during their coverage gap period,
as a condition for the manufacturer’s outpatient drugs to be
covered under Medicare Part D; imposed a non-deductible annual fee
on pharmaceutical manufacturers or importers who sell certain
“branded prescription drugs” to specified federal government
programs; implemented a new methodology by which rebates owed by
manufacturers under the Medicaid Drug Rebate Program are calculated
for drugs
70
that are inhaled, infused, instilled, implanted, or injected;
expanded eligibility criteria for Medicaid programs; creates a new
Patient-Centered Outcomes Research Institute to oversee, identify
priorities in, and conduct comparative clinical effectiveness
research, along with funding for such research; and established a
Center for Medicare Innovation at the Centers for Medicare &
Medicaid Services (CMS) to test innovative payment and service
delivery models to lower Medicare and Medicaid spending,
potentially including prescription drug spending.
Since its enactment, there have been judicial, executive and
political challenges to certain aspects of the ACA. On June 17,
2021, the US Supreme Court dismissed a challenge on procedural
grounds that argued the ACA is unconstitutional in its entirety
because the “individual mandate” was repealed by Congress. Thus,
the ACA will remain in effect in its current form. Prior to the US
Supreme Court ruling, President Biden issued an executive order
that initiated initiate a special enrollment period from February
15, 2021 through August 15, 2021 for purposes of obtaining health
insurance coverage through the ACA marketplace. The executive order
also instructed certain governmental agencies to review and
reconsider their existing policies and rules that limit access to
healthcare, including among others, reexamining Medicaid
demonstration projects and waiver programs that include work
requirements, and policies that create unnecessary barriers to
obtaining access to health insurance coverage through Medicaid or
the ACA. It is unclear how other healthcare reform measures of the
Biden administration will impact our business.
In addition, other legislative changes have been proposed and
adopted since the ACA was enacted. These changes included aggregate
reductions to Medicare payments to providers of 2% per fiscal year,
which went into effect on April 1, 2013 and, due to subsequent
legislative amendments to the statute, will remain in effect
through 2030, with the exception of a temporary suspension from May
1, 2020 through March 31, 2022, unless additional Congressional
action is taken. On January 2, 2013, the American Taxpayer Relief
Act of 2012 was signed into law, which, among other things, reduced
Medicare payments to several providers, including hospitals, and
increased the statute of limitations period for the government to
recover overpayments to providers from three to five
years.
Moreover, there has recently been heightened governmental scrutiny
over the manner in which manufacturers set prices for their
marketed products, which has resulted in several Congressional
inquiries and proposed and enacted federal and state legislation
designed to, among other things, bring more transparency to product
pricing, review the relationship between pricing and manufacturer
patient programs, and reform government program reimbursement
methodologies for pharmaceutical products. Further, we expect that
additional healthcare reform measures will be adopted in the
future, particularly in light of the new presidential
administration.
Individual states in the United States have also become
increasingly active in implementing regulations designed to control
pharmaceutical product pricing, including price or patient
reimbursement constraints, discounts, restrictions on certain
product access and marketing cost disclosure and transparency
measures, and, in some cases, designed to encourage importation
from other countries and bulk purchasing. In addition, regional
healthcare authorities and individual hospitals are increasingly
using bidding procedures to determine which drugs and suppliers
will be included in their healthcare programs Furthermore, there
has been increased interest by third party payors and governmental
authorities in reference pricing systems and publication of
discounts and list prices.
EU drug regulation
In order to market any product outside of the United States, we
would need to comply with numerous and varying regulatory
requirements of other countries and jurisdictions regarding
quality, safety and efficacy and governing, among other things,
clinical trials, marketing authorization, commercial sales and
distribution of our products. Whether or not we obtain FDA approval
for a product, we would need to obtain the necessary approvals by
the comparable foreign regulatory authorities before we can
commence clinical trials or marketing of the product in foreign
countries and jurisdictions such as in China and Japan. Although
many of the issues discussed above with respect to the United
States apply similarly in the context of the European Union (EU),
the approval process varies between countries and jurisdictions and
can involve additional product testing and additional
administrative review periods. The time required to obtain approval
in other countries and jurisdictions might differ from and be
longer than that required to obtain FDA approval. Regulatory
approval in one country or jurisdiction does not ensure regulatory
approval in another, but a failure or delay in obtaining regulatory
approval in one country or jurisdiction may negatively impact the
regulatory process in others. Failure to comply with applicable
foreign regulatory requirements, may be subject to, among other
things, fines, suspension or withdrawal of regulatory approvals,
product recalls, seizure of products, operating restrictions and
criminal prosecution.
Non-clinical studies and clinical trials
Similarly to the United States, the various phases of non-clinical
and clinical research in the EU are subject to significant
regulatory controls.
71
Non-clinical studies are performed to demonstrate the health or
environmental safety of new chemical or biological substances.
Non-clinical studies must be conducted in compliance with the
principles of good laboratory practice (GLP) as set forth in EU
Directive 2004/10/EC. In particular, non-clinical studies, both in
vitro and in vivo, must be planned, performed, monitored, recorded,
reported and archived in accordance with the GLP principles, which
define a set of rules and criteria for a quality system for the
organizational process and the conditions for non-clinical studies.
These GLP standards reflect the Organization for Economic
Co-operation and Development requirements.
Clinical trials of medicinal products in the EU must be conducted
in accordance with EU and national regulations and the
International Conference on Harmonization (ICH) guidelines on GCP
as well as the applicable regulatory requirements and the ethical
principles that have their origin in the Declaration of Helsinki.
Additional GCP guidelines from the European Commission, focusing in
particular on traceability, apply to clinical trials of advanced
therapy medicinal products. If the sponsor of the clinical trial is
not established within the EU, it must appoint an entity within the
EU to act as its legal representative. The sponsor must take out a
clinical trial insurance policy, and in most EU member states, the
sponsor is liable to provide ‘no fault’ compensation to any study
subject injured in the clinical trial.
Certain countries outside of the United States, including the EU,
have a similar process that requires the submission of a clinical
study application (CTA) much like the IND prior to the commencement
of human clinical studies. A CTA must be submitted to each
country’s national health authority and an independent ethics
committee, much like the FDA and the IRB, respectively. Once the
CTA is approved by the national health authority and the ethics
committee has granted a positive opinion in relation to the conduct
of the trial in the relevant member state(s), in accordance with a
country’s requirements, clinical study development may
proceed.
The CTA must include, among other things, a copy of the trial
protocol and an investigational medicinal product dossier
containing information about the manufacture and quality of the
medicinal product under investigation. Currently, CTAs must be
submitted to the competent authority in each EU member state in
which the trial will be conducted. Under the new Regulation on
Clinical Trials, which is currently expected to become applicable
by early 2022, there will be a centralized application procedure
where one national authority takes the lead in reviewing the
application and the other national authorities have only a limited
involvement. Any substantial changes to the trial protocol or other
information submitted with the CTA must be notified to or approved
by the relevant competent authorities and ethics committees.
Medicines used in clinical trials must be manufactured in
accordance with good manufacturing practice (GMP). Other national
and EU-wide regulatory requirements also apply.
Marketing Authorizations
To market a medicinal product in the EU and in many other foreign
jurisdictions, we must obtain separate regulatory approvals. More
concretely, in the EU, medicinal product candidates can only be
commercialized after obtaining a Marketing Authorization (MA). To
obtain regulatory approval of an investigational medicinal product
under EU regulatory systems, we must submit a marketing
authorization application (MAA.) The process for doing this
depends, among other things, on the nature of the medicinal
product. There are two types of MAs:
•
the “Union MA”, which is issued by the European Commission through
the Centralized Procedure, based on the opinion of the Committee
for Medicinal Products for Human Use (CHMP) of the European
Medicines Agency (EMA) and which is valid throughout the entire
territory of the EU. The Centralized Procedure is mandatory for
certain types of products, such as: (i) medicinal products derived
from biotechnology medicinal products, (ii) designated orphan
medicinal products, (iii) advanced therapy products (such as gene
therapy, somatic cell therapy or tissue-engineered medicines), and
(iv) medicinal products containing a new active substance indicated
for the treatment certain diseases, such as HIV/AIDS, cancer,
neurodegenerative diseases, diabetes, other auto-immune and viral
diseases. The Centralized Procedure is optional for products
containing a new active substance not yet authorized in the EU, or
for products that constitute a significant therapeutic, scientific
or technical innovation or that the granting of authorization would
be in the interest of public health in the EU; and
•
“National MAs”, which are issued by the competent authorities of
the EU member states and only cover their respective territory, are
available for products not falling within the mandatory scope of
the Centralized Procedure. Where a product has already been
authorized for marketing in an EU member state, this National MA
can be recognized in another member state through the Mutual
Recognition Procedure. If the product has not received a National
MA in any member state at the time of application, it can be
approved simultaneously in various member states through the
Decentralized Procedure. Under the Decentralized Procedure an
identical dossier is submitted to the competent authorities of each
of the member states in which the MA is sought, one of which is
selected by the applicant as the Reference member
state.
72
Under the above-described procedures, in order to grant the MA, the
EMA or the competent authorities of the EU member states make an
assessment of the risk-benefit balance of the product on the basis
of scientific criteria concerning its quality, safety and
efficacy.
Under the Centralized Procedure, the maximum timeframe for the
evaluation of a MAA by the EMA is 210 days. Where there is a major
public health interest and an unmet medical need for a product, the
CHMP may perform an accelerated review of a MA in no more than 150
days (not including clock stops). Innovative products that target
an unmet medical need and are expected to be of major public health
interest may be eligible for a number of expedited development and
review programs, such as the PRIME scheme, which provides
incentives similar to the breakthrough therapy designation in the
US PRIME is a voluntary scheme aimed at enhancing the EMA’s support
for the development of medicines that target unmet medical needs.
It is based on increased interaction and early dialogue with
companies developing promising medicines, to optimize their product
development plans and speed up their evaluation to help them reach
patients earlier. Product developers that benefit from PRIME
designation can expect to be eligible for accelerated assessment
but this is not guaranteed. The benefits of a PRIME designation
include the appointment of a CHMP rapporteur before submission of a
MAA, early dialogue and scientific advice at key development
milestones, and the potential to qualify products for accelerated
review earlier in the application process.
MAs have an initial duration of five years. After these five years,
the authorization may be renewed for an unlimited period on the
basis of a reevaluation of the risk-benefit balance, unless the EMA
decides, on justified grounds relating to pharmacovigilance, to
mandate one additional five-year renewal period.
Data and marketing exclusivity
The EU also provides opportunities for market exclusivity. Upon
receiving MA, new chemical entity, or reference product candidates,
generally receive eight years of data exclusivity and an additional
two years of market exclusivity. If granted, the data exclusivity
period prevents generic or biosimilar applicants from relying on
the pre-clinical and clinical trial data contained in the dossier
of the reference product when applying for a generic or biosimilar
MA in the EU during a period of eight years from the date on which
the reference product was first authorized in the EU. The market
exclusivity period prevents a successful generic or biosimilar
applicant from commercializing its product in the EU until 10 years
have elapsed from the initial authorization of the reference
product in the EU. The overall 10-year market exclusivity period
can be extended to a maximum of eleven years if, during the first
eight years of those 10 years, the MA holder obtains an
authorization for one or more new therapeutic indications which,
during the scientific evaluation prior to their authorization, are
held to bring a significant clinical benefit in comparison with
existing therapies. However, there is no guarantee that a product
will be considered by the EU’s regulatory authorities to be a new
chemical entity, and products may not qualify for data
exclusivity.
Pediatric Development
In the EU, MAAs for new medicinal products candidates have to
include the results of trials conducted in the pediatric
population, in compliance with a pediatric investigation plan (PIP)
agreed with the EMA’s Pediatric Committee (PDCO). The PIP sets out
the timing and measures proposed to generate data to support a
pediatric indication of the drug for which MA is being sought. The
PDCO can grant a deferral of the obligation to implement some or
all of the measures of the PIP until there is sufficient data to
demonstrate the efficacy and safety of the product in adults.
Further, the obligation to provide pediatric clinical trial data
can be waived by the PDCO when these data is not needed or
appropriate because the product is likely to be ineffective or
unsafe in children, the disease or condition for which the product
is intended occurs only in adult populations, or when the product
does not represent a significant therapeutic benefit over existing
treatments for pediatric patients. Once the MA is obtained in all
EU Member States and study results are included in the product
information, even when negative, the product is eligible for six
months’ supplementary protection certificate extension (if any is
in effect at the time of authorization).
Post-Approval Requirements
Similar to the United States, both MA holders and manufacturers of
medicinal products are subject to comprehensive regulatory
oversight by the EMA, the European Commission and/or the competent
regulatory authorities of the member states. The holder of a MA
must establish and maintain a pharmacovigilance system and appoint
an individual qualified person for pharmacovigilance who is
responsible for oversight of that system. Key obligations include
expedited reporting of suspected serious adverse reactions and
submission of periodic safety update reports (PSURs).
73
All new MAA must include a risk management plan (RMP) describing
the risk management system that the company will put in place and
documenting measures to prevent or minimize the risks associated
with the product. The regulatory authorities may also impose
specific obligations as a condition of the MA. Such
risk-minimization measures or post-authorization obligations may
include additional safety monitoring, more frequent submission of
PSURs, or the conduct of additional clinical trials or
post-authorization safety studies.
The advertising and promotion of medicinal products is also subject
to laws concerning promotion of medicinal products, interactions
with physicians, misleading and comparative advertising and unfair
commercial practices. All advertising and promotional activities
for the product must be consistent with the approved summary of
product characteristics, and therefore all off-label promotion is
prohibited. Direct-to-consumer advertising of prescription
medicines is also prohibited in the EU. Although general
requirements for advertising and promotion of medicinal products
are established under EU directives, the details are governed by
regulations in each member state and can differ from one country to
another.
The aforementioned EU rules are generally applicable in the
European Economic Area (EEA) which consists of the 27 EU member
states plus Norway, Liechtenstein and Iceland.
For other countries outside of the EU, such as countries in Latin
America or Asia (e.g., China and Japan), the requirements governing
the conduct of clinical studies, product licensing, pricing and
reimbursement vary from country to country. In all cases, again,
the clinical studies are conducted in accordance with GCP and the
applicable regulatory requirements and the ethical principles that
have their origin in the Declaration of Helsinki. If we fail to
comply with applicable foreign regulatory requirements, we may be
subject to, among other things, fines, suspension or withdrawal of
regulatory approvals, product recalls, seizure of products,
operating restrictions and criminal prosecution.
Data Privacy and Security Laws
Numerous state, federal and foreign laws, regulations and standards
govern the collection, use, access to, confidentiality and security
of health-related and other personal information, and could apply
now or in the future to our operations or the operations of our
partners. In the United States, numerous federal and state laws and
regulations, including data breach notification laws, health
information privacy and security laws and consumer protection laws
and regulations govern the collection, use, disclosure, and
protection of health-related and other personal information. In
addition, certain foreign laws govern the privacy and security of
personal data, including health-related data. For example, the GDPR
imposes strict requirements for processing the personal data of
individuals within the EEA. Companies that must comply with the
GDPR face increased compliance obligations and risk, including more
robust regulatory enforcement of data protection requirements and
potential fines for noncompliance of up to €20 million or 4% of the
annual global revenues of the noncompliant company, whichever is
greater. Further, from January 1, 2021, companies have had to
comply with the GDPR and also the UK GDPR, which, together with the
amended UK Data Protection Act 2018, retains the GDPR in UK
national law. The UK GDPR mirrors the fines under the GDPR, i.e.,
fines up to the greater of €20 million (£17.5 million) or 4% of
global turnover. Privacy and security laws, regulations, and other
obligations are constantly evolving, may conflict with each other
to complicate compliance efforts, and can result in investigations,
proceedings, or actions that lead to significant civil and/or
criminal penalties and restrictions on data processing.
Japanese drug regulation
Non-clinical studies and clinical trials
Being a member of the International Conference on Harmonization
(ICH), Japan has pharmaceutical regulations fundamentally similar
to those of the United States or EU.
Non-clinical studies are performed to demonstrate the health safety
of new chemical or biological substances. Non-clinical studies must
be conducted in compliance with the principles of Japanese good
laboratory practice (GLP) which reflect the Organization for
Economic Co-operation and Development requirements. Currently,
Japan and EU have a mutual recognition agreement for GLP, and data
generated compliant with EU requirements will be accepted by the
Japanese authorities. There is no similar agreement with the United
States.
Clinical trials of medicinal products in Japan must be conducted in
accordance with Japanese regulations based on ICH guidelines
governing good clinical practices (GCP). They focus on ethics of
the clinical trial and protection of the privacy of the trial
subjects. If the sponsor of the clinical trial is not established
within Japan, it must appoint an entity within the country to act
as its caretaker who should be authorized to act on the sponsor’s
behalf. The sponsor must take out a clinical trial insurance
policy, and, according to the industry agreement, should put in
place a common compensation policy for the injuries from the
trial.
74
Prior to the commencement of human clinical studies, the sponsor
must complete evaluation of the safety of the investigative
product, and submit a clinical trial notification and the protocol
to the authorities in advance, upon agreement of the IRB of the
participating institutions. When the authorities do not comment on
the notification, the sponsor may proceed with the clinical
trial.
Any substantial changes to the trial protocol or other information
submitted must be cleared by the IRB and notified to the
authorities. Medicines used in clinical trials must be manufactured
in accordance with good manufacturing practice (GMP).
Product approval
To market a medicinal product in Japan, we must obtain regulatory
approval. To obtain regulatory approval of an investigational
medicinal product, we must submit a new drug application. The
process for doing this depends, among other things, on the nature
of the medicinal product and there are currently a few different
pathways for approval. If the product is designed for treating
certain “difficult diseases” or those whose patient size is
limited, we may be able to obtain designation as an orphan drug
product if it demonstrates unique therapeutic value. Approval
application for such designated orphan products will be processed
on an expedited basis and the authorities’ requirement for clinical
data will be much limited. Separately, the latest amendment to the
law introduced separate pathways for: (i) truly innovative products
with a unique mode of action, and (ii) those which will satisfy
unmet medical needs. These products will also be processed on an
expedited basis.
The evaluation of applications will be based on an assessment of
the risk-benefit balance of the product on the basis of scientific
criteria concerning its quality, safety and efficacy. Once the
review organization completes its review task, the matter will be
considered by the advisory committee of experts, and the government
will grant approval upon positive recommendation from the
committee.
The volume and quality of the clinical data will be the key
determinant of the approval decision. Clinical trial data generated
overseas will be accepted as part of the data package consistent
with the ICH recommendation. Typically, a limited dose response
clinical trial for Japanese subjects is required to ensure that
data are extrapolatable for the Japanese population. In a more
recent development, the authorities encourage manufacturers to
organize an international joint clinical trial with some Japanese
participation under a joint protocol, to expedite the clinical
trial process. Regulatory approval does not expire.
Licensing requirement
Separate from the approval requirement, it is also mandatory to
possess a distribution license of an appropriate class for the
manufacturer to commercially distribute the product in Japan.
Non-Japanese companies who possess only the product approval may
designate an appropriate license holder in Japan to commercially
distribute the product, rather than distributing it on its own. The
license is valid for 5 years.
Facilities
Our corporate headquarters are located in San Diego, California,
where we currently lease approximately 16,153 square feet of office
and laboratory space pursuant to a lease that expires April 1,
2022, with the option to holdover the lease month-to-month
following such date. We plan to exercise this holdover option until
we move into our new corporate headquarters, which is anticipated
to occur in the second quarter of 2022. In September 2020, we
entered into a lease of approximately 59,407 square feet of office
and laboratory space in a facility in San Diego, California that is
currently under construction, and scheduled to be completed in the
second quarter of 2022. In March 2021, we amended this lease to
include an additional 18,421 square feet of office space, resulting
in 77,828 square feet of total leased space within this facility.
The lease has an initial term of 10.5 years and commenced in August
2021. We believe our existing facilities are adequate to meet our
current business requirements for the near term, and that
additional space will be available on commercially reasonable
terms, if required.
75
Employees
As of February 28, 2022, we had 123 full-time employees (FTEs), 50
of whom have doctorate degrees. Of our FTEs, 87 are engaged in
research and development activities, and 36 are engaged in general
and administrative activities. Substantially all of our employees
are located in San Diego County, California. None of our employees
are represented by labor unions or covered by collective bargaining
units. We consider our relationship with our employees to be
good.
Our human resources objectives include, as applicable, identifying,
recruiting, retaining, incentivizing, and integrating our existing
and additional employees. The principal purposes of our equity
incentive plans are to attract, retain, and motivate selected
employees, consultants, and directors through the granting of
stock-based compensation awards.
Corporate Information
We were incorporated under the laws of the State of Delaware on
July 2, 2018 as Erasca, Inc. Our principal executive offices are
located at 10835 Road to the Cure, Suite 140, San Diego, California
92121, and our telephone number is 858-465-6511. Our website
address is www.erasca.com. Our website and the information
contained on, or that can be accessed through, the website will not
be deemed to be incorporated by reference in, and are not
considered part of, this Annual Report on Form 10-K.
In July 2021, we completed our IPO pursuant to which we issued and
sold 21,562,500 shares of common stock at a public offering price
of $16.00 per share, resulting in net proceeds of $317 million,
after deducting underwriting discounts and commissions and other
offering costs.
We are an "emerging growth company" as defined in the Jumpstart Our
Business Startups Act of 2012. We will remain an emerging growth
company until the earlier of: (i) the last day of the fiscal year:
(a) following the fifth anniversary of the completion of the IPO,
(b) in which we have total annual gross revenue of at least $1.07
billion, or (c) in which we are deemed to be a large accelerated
filer, which means the market value of our common stock that is
held by non-affiliates exceeds $700.0 million as of the prior June
30th, and (ii) the date on which we have issued more than $1.0
billion in nonconvertible debt during the prior three-year
period.
Available Information
Our website address is www.erasca.com. Our Annual Reports on Form
10-K, Quarterly Reports on Form 10-Q, Current Reports on Form 8-K,
including exhibits, proxy and information statements and amendments
to those reports filed or furnished pursuant to Sections 13(a), 14,
and 15(d) of the Exchange Act are available through the “Investors”
portion of our website free of charge as soon as reasonably
practicable after we electronically file such material with, or
furnish it to, the SEC. In addition, our filings with the SEC may
be accessed through the SEC’s Interactive Data Electronic
Applications system at www.sec.gov. All statements made in any of
our securities filings, including all forward-looking statements or
information, are made as of the date of the document in which the
statement is included, and we do not assume or undertake any
obligation to update any of those statements or documents unless we
are required to do so by law.
We use the “Investors” portion of our website as a means of
disclosing material non-public information and for complying with
our disclosure obligations under Regulation FD. Investors should
monitor such website, in addition to following our press releases,
SEC filings and public conference calls and webcasts. Information
relating to our corporate governance is also included on our
website. The information in or accessible through the SEC and our
website are not incorporated into, and are not considered part of,
this Annual Report on Form 10-K.
76
Item 1A. Risk
Factors.
Investing in our common stock involves a high degree of risk. You
should carefully consider the risks and uncertainties described
below, as well as the other information in this Annual Report on
Form 10-K, including our consolidated financial statements and the
related notes included elsewhere in this Annual Report on Form 10-K
and “Management’s Discussion and Analysis of Financial Condition
and Results of Operations,” before making investment decisions
regarding our common stock. The occurrence of any of the events or
developments described below could harm our business, financial
condition, results of operations and prospects. In such an event,
the market price of our common stock could decline and you may lose
all or part of your investment. The risks described below are not
the only ones we face. Additional risks and uncertainties not
presently known to us or that we currently deem immaterial also may
impair our business operations.
Risks related to our limited operating history, financial position
and need for additional capital
We have a limited operating history, have incurred significant
operating losses since our inception and expect to incur
significant losses for the foreseeable future. We may never
generate any revenue or become profitable or, if we achieve
profitability, we may not be able to sustain it.
We are a clinical-stage biopharmaceutical company with a limited
operating history upon which you can evaluate our business and
prospects. We commenced operations in 2018, and to date, we have
focused primarily on organizing and staffing our company, business
planning, raising capital, identifying, acquiring and in-licensing
our product candidates, establishing our intellectual property
portfolio, conducting research, preclinical studies and clinical
trials, establishing arrangements with third parties for the
manufacture of our product candidates and related raw materials,
and providing general and administrative support for these
operations. Our scientific approach to the discovery and
development of product candidates is unproven, and we do not know
whether we will be able to develop or obtain regulatory approval
for any products of commercial value. In addition, we only have
three product candidates, ERAS-007, ERAS-601, and ERAS-801, in
early clinical development, and our other product candidates remain
in the preclinical or discovery stage. We have not yet completed
any later-stage, large-scale or pivotal clinical trials, obtained
regulatory approvals, manufactured a commercial-scale product, or
arranged for a third party to do so on our behalf, or conducted
sales and marketing activities necessary for successful product
commercialization. Consequently, any predictions made about our
future success or viability may not be as accurate as they could be
if we had a history of successfully developing and commercializing
biopharmaceutical products.
We have incurred significant operating losses since our inception
and expect to incur significant losses for the foreseeable future.
We do not have any products approved for sale and have not
generated any revenue since our inception. If we are unable to
successfully develop and obtain requisite approval for our product
candidates, we may never generate any revenue. Our net losses were
$122.8 million and $101.7 million for the years ended December 31,
2021 and 2020, respectively. As of December 31, 2021, we had an
accumulated deficit of $238.2 million. Substantially all of our
losses have resulted from expenses incurred in connection with our
research and development programs and from general and
administrative costs associated with our operations. All of our
product candidates will require substantial additional development
time and resources before we would be able to apply for or receive
regulatory approvals and begin generating revenue from product
sales. We expect to continue to incur losses for the foreseeable
future, and we anticipate these losses will increase substantially
as we continue our development of, seek regulatory approval for and
potentially commercialize any of our product candidates and seek to
identify, assess, acquire, in-license or develop additional product
candidates.
To become and remain profitable, we must succeed in developing and
eventually commercializing products that generate significant
revenue. This will require us to be successful in a range of
challenging activities, including completing clinical trials and
preclinical studies of our product candidates, discovering,
acquiring or in-licensing additional product candidates, obtaining
regulatory approval for these product candidates, and
manufacturing, marketing, and selling any products for which we may
obtain regulatory approval. We are only in the preliminary stages
of most of these activities. We may never succeed in these
activities and, even if we do, may never generate revenue that is
significant enough to achieve profitability. In addition, we have
not yet demonstrated an ability to successfully overcome many of
the risks and uncertainties frequently encountered by companies in
new and rapidly evolving fields, particularly in the
biopharmaceutical industry. Because of the numerous risks and
uncertainties associated with biopharmaceutical product
development, we are unable to accurately predict the timing or
amount of increased expenses or when, or if, we will be able to
achieve profitability. Even if we do achieve profitability, we may
not be able to sustain or increase profitability on a quarterly or
annual basis. Our failure to become and remain profitable may have
an adverse effect on the value of our company and could impair our
ability to raise capital, expand our business, maintain our
research and development
77
efforts, diversify our product candidates, or even continue our
operations. A decline in the value of our company could also cause
you to lose all or part of your investment.
We will require substantial additional capital to finance our
operations, and a failure to obtain this necessary capital when
needed on acceptable terms, or at all, could force us to delay,
limit, reduce or terminate our development programs,
commercialization efforts or other operations.
The development of biopharmaceutical product candidates is
capital-intensive. Our operations have consumed substantial amounts
of cash since inception. We expect our expenses to increase in
connection with our ongoing activities, particularly as we conduct
our ongoing and planned clinical trials and preclinical studies,
and seek regulatory approval for our current product candidates and
any future product candidates we may develop or otherwise acquire.
In addition, as our product candidates progress through development
and toward commercialization, we will need to make milestone
payments to the licensors and other third parties from whom we have
in-licensed or acquired our product candidates, including ERAS-007
and ERAS-601. If we obtain regulatory approval for any of our
product candidates, we also expect to incur significant
commercialization expenses related to product manufacturing,
marketing, sales, and distribution. Because the outcome of any
clinical trial or preclinical study is highly uncertain, we cannot
reasonably estimate the actual amounts necessary to successfully
complete the development and commercialization of our product
candidates. Furthermore, we expect to incur additional costs
associated with operating as a public company. Accordingly, we will
need to obtain substantial additional funding in connection with
our continuing operations. If we are unable to raise capital when
needed or on attractive terms, we could be forced to delay, reduce
or eliminate our research and development programs or any future
commercialization efforts.
Based on our current operating plan, we believe that our existing
cash, cash equivalents and investments will be sufficient to fund
our operations into 2024. We have based these estimates on
assumptions that may prove to be wrong, and we could use our
capital resources sooner than we currently expect. Our operating
plans and other demands on our cash resources may change as a
result of many factors currently unknown to us, and we may need to
seek additional funds sooner than planned, through public or
private equity or debt financings or other capital sources,
including potential collaborations, licenses and other similar
arrangements. In addition, we may seek additional capital due to
favorable market conditions or strategic considerations even if we
believe we have sufficient funds for our current or future
operating plans. Attempting to secure additional financing may
divert our management from our day-to-day activities, which may
adversely affect our ability to develop our product
candidates.
Our future capital requirements will depend on many factors,
including, but not limited to:
•
the type, number, scope, progress, expansions, results, costs and
timing of discovery, preclinical studies and clinical trials of our
product candidates which we are pursuing or may choose to pursue in
the future, including the costs of any third-party products used in
our combination clinical trials that are not covered by such third
party or other sources;
•
the costs and timing of manufacturing for our product candidates,
including commercial manufacturing if any product candidate is
approved;
•
the costs, timing and outcome of regulatory review of our product
candidates;
•
the costs of obtaining, maintaining and enforcing our patents and
other intellectual property rights;
•
our efforts to enhance operational systems and hire additional
personnel to satisfy our obligations as a public company, including
enhanced internal controls over financial reporting;
•
the costs associated with hiring additional personnel and
consultants as our preclinical and clinical activities
increase;
•
the timing and amount of the milestone or other payments we must
make to the licensors and other third parties from whom we have
in-licensed our acquired our product candidates;
•
the costs and timing of establishing or securing sales and
marketing capabilities if any product candidate is
approved;
•
our ability to achieve sufficient market acceptance, coverage and
adequate reimbursement from third-party payors and adequate market
share and revenue for any approved products;
•
patients’ willingness to pay out-of-pocket for any approved
products in the absence of coverage and/or adequate reimbursement
from third-party payors;
78
•
any delays and cost increases that result from the COVID-19
pandemic;
•
the terms and timing of establishing and maintaining
collaborations, licenses and other similar arrangements;
and
•
costs associated with any products or technologies that we may
in-license or acquire.
Conducting clinical trials and preclinical studies and identifying
potential product candidates is a time-consuming, expensive and
uncertain process that takes years to complete, and we may never
generate the necessary data or results required to obtain
regulatory approval and commercialize our product candidates. In
addition, our product candidates, if approved, may not achieve
commercial success. Our commercial revenue, if any, will be derived
from sales of products that we do not expect to be commercially
available for many years, if at all.
Accordingly, we will need to continue to rely on additional
financing to achieve our business objectives. Adequate additional
financing may not be available to us on acceptable terms, or at
all.
Raising additional capital may cause dilution to our stockholders,
restrict our operations or require us to relinquish rights to our
technologies or product candidates.
Until such time, if ever, as we can generate substantial product
revenue, we expect to finance our cash needs through equity
offerings, debt financings, or other capital sources, including
potential collaborations, licenses and other similar arrangements.
We do not have any committed external source of funds. To the
extent that we raise additional capital through the sale of equity
or convertible debt securities, your ownership interest will be
diluted, and the terms of these securities may include liquidation
or other preferences that adversely affect your rights as a common
stockholder. Debt financing and preferred equity financing, if
available, may involve agreements that include covenants limiting
or restricting our ability to take specific actions, such as
incurring additional debt, making capital expenditures or declaring
dividends. Such restrictions could adversely impact our ability to
conduct our operations and execute our business plan.
If we raise additional funds through future collaborations,
licenses and other similar arrangements, we may have to relinquish
valuable rights to our future revenue streams, research programs or
product candidates, or grant licenses on terms that may not be
favorable to us and/or that may reduce the value of our common
stock. If we are unable to raise additional funds through equity or
debt financings or other arrangements when needed or on terms
acceptable to us, we would be required to delay, limit, reduce, or
terminate our product development or future commercialization
efforts or grant rights to develop and market product candidates
that we would otherwise prefer to develop and market
ourselves.
Risks related to the discovery, development and regulatory approval
of our product candidates
We are early in our development efforts with three product
candidates in early clinical development. All of our other
development programs are still in the preclinical or discovery
stage. If we are unable to successfully develop, obtain regulatory
approval and ultimately commercialize any of our current or future
product candidates, or experience significant delays in doing so,
our business will be materially harmed.
We are early in our development efforts and have three product
candidates, ERAS-007, ERAS-601, and ERAS-801, in early clinical
development. All of our other programs are still in the preclinical
or discovery stage. Our ability to generate product revenue, which
we do not expect will occur for many years, if ever, will depend
heavily on the successful development and eventual
commercialization of our product candidates. The success of our
product candidates will depend on several factors, including the
following:
•
initiation and successful enrollment of clinical trials and timely
completion of clinical trials and preclinical studies with
favorable results;
•
acceptance of INDs by the FDA, or of similar regulatory submissions
by comparable foreign regulatory authorities for the conduct of
clinical trials of our product candidates and our proposed design
of future clinical trials;
•
the frequency and severity of adverse events in clinical
trials;
•
maintaining and establishing relationships with contract research
organizations (CROs) and clinical sites for the clinical
development of our product candidates both in the United States and
internationally;
•
demonstrating the safety, purity, potency and efficacy of our
product candidates to the satisfaction of applicable regulatory
authorities;
79
•
receipt of marketing approvals from applicable regulatory
authorities, including NDAs and BLAs from the FDA and maintaining
such approvals;
•
making arrangements with our third-party manufacturers for, or
establishing, commercial manufacturing capabilities;
•
establishing sales, marketing and distribution capabilities and
launching commercial sales of our products, if and when approved,
whether alone or in collaboration with others;
•
establishing and maintaining patent and trade secret protection or
regulatory exclusivity for our product candidates;
•
maintaining an acceptable safety profile of our products following
approval, if any; and
•
maintaining and growing an organization of people who can develop
and commercialize our products and technology.
If we are unable to develop, obtain regulatory approval for, or, if
approved, successfully commercialize our product candidates, we may
not be able to generate sufficient revenue to continue our
business.
Our scientific approach to the discovery and development of product
candidates is unproven, and we do not know whether we will be able
to develop any products of commercial value, or if competing
approaches will limit the commercial value of our product
candidates.
The success of our business depends primarily upon our ability to
identify, develop and commercialize products based on our
scientific approach, which is singularly focused on shutting down
the RAS/MAPK pathway, a novel and unproven approach. While we have
had favorable preclinical study results for certain of our
development programs, we have not yet succeeded and may not succeed
in demonstrating efficacy and safety for any product candidates in
clinical trials or in obtaining marketing approvals from the FDA or
other regulatory authorities or in commercializing such product
candidates. Our lead product candidates, ERAS-007 and ERAS-601, as
well as our ERAS-801 product candidate, are in early clinical
development and, as an organization, we have not completed any
clinical trials for any of our product candidates. In addition,
while we believe our pipeline will yield multiple additional INDs
for our development programs in the future, we may not be
successful in our discovery efforts, and even if successful, we may
not be able to submit INDs and have such INDs accepted to enable us
to commence clinical trials on the timelines we expect, if at all.
Our research methodology and scientific approach may be
unsuccessful in identifying additional product candidates, and any
product candidates may be shown to have harmful side effects or may
have other characteristics that may necessitate additional clinical
testing, or make the product candidates unmarketable or unlikely to
receive marketing approval. In particular, using multiple agents to
shut down multiple nodes of the RAS/MAPK pathway simultaneously is
a novel approach that may have unexpected consequences, including
adverse events that preclude successful development and approval of
our product candidates. Further, because all of our current product
candidates and development programs are based on the RAS/MAPK
pathway, adverse developments with respect to one of our programs
may have a significant adverse impact on the actual or perceived
likelihood of success and value of our other programs.
In addition, the biotechnology and biopharmaceutical industries are
characterized by rapidly advancing technologies. Our future success
will depend in part on our ability to maintain a competitive
position with our scientific approach. If we fail to stay at the
forefront of technological change in utilizing our approach to
create and develop product candidates, we may be unable to compete
effectively. Our competitors may render our approach obsolete, or
limit the commercial value of our products or product candidates by
advances in existing technological approaches or the development of
new or different approaches, potentially eliminating the advantages
in our drug discovery process that we believe we derive from our
approach. By contrast, adverse developments with respect to other
companies that attempt to use a similar approach to our approach
may adversely impact the actual or perceived value and potential of
our product candidates.
If any of these events occur, we may be forced to delay, modify, or
abandon our development efforts for a program or programs, which
would have a material adverse effect on our business and could
potentially cause us to cease operations.
80
Clinical and preclinical development involves a lengthy and
expensive process with an uncertain outcome, and the results of
preclinical studies and early clinical trials are not necessarily
predictive of future results. Any of our product candidates may not
have favorable results in clinical trials, if any, or receive
regulatory approval on a timely basis, if at all.
Clinical and preclinical development is expensive and can take many
years to complete, and its outcome is inherently uncertain. We
cannot guarantee that any clinical trials or preclinical studies
will be conducted as planned or completed on schedule, if at all,
and failure can occur at any time during the preclinical study or
clinical trial process, including due to factors that are beyond
our control. Further, we may not be able to meet expected
timeframes for data readouts, such as those for our FLAGSHP-1
clinical trial or HERKULES series of clinical trials. Despite
promising preclinical or clinical results, any product candidate
can unexpectedly fail at any stage of preclinical or clinical
development. The historical failure rate for product candidates in
our industry is high.
The results from preclinical studies or clinical trials of a
product candidate or a competitor’s product candidate in the same
class may not predict the results of later clinical trials of our
product candidate, and interim, topline, or preliminary results of
a clinical trial are not necessarily indicative of final results.
Product candidates in later stages of clinical trials may fail to
show the desired safety and efficacy characteristics despite having
progressed through preclinical studies and initial clinical trials.
For example, while a Phase 1 clinical trial of ERAS-007 was
completed prior to our acquisition of this product candidate and
while we have completed IND-enabling preclinical studies of
ERAS-601 and ERAS-801, we do not know whether they or our other
potential product candidates will perform in ongoing or future
clinical trials as they have performed in these prior trials and
studies. It is not uncommon to observe results in clinical trials
that are unexpected based on preclinical studies and early clinical
trials, and many product candidates fail in clinical trials despite
very promising early results. We are currently conducting
IND-enabling preclinical studies for ERAS-3490. If unexpected
observations or toxicities are observed in these studies, or in
future IND-enabling studies for any of our other development
programs, such results may delay or prevent the initiation of
clinical trials for such development programs. Moreover,
preclinical and clinical data may be susceptible to varying
interpretations and analyses. A number of companies in the
biopharmaceutical and biotechnology industries have suffered
significant setbacks in clinical development even after achieving
promising results in earlier studies.
For the foregoing reasons, we cannot be certain that our ongoing
and planned clinical trials and preclinical studies will be
successful. Any safety concerns observed in any one of our clinical
trials in our targeted indications could limit the prospects for
regulatory approval of our product candidates in those and other
indications, which could have a material adverse effect on our
business, financial condition and results of operations.
Any difficulties or delays in the commencement or completion, or
termination or suspension, of our current or planned clinical
trials could result in increased costs to us, delay or limit our
ability to generate revenue and adversely affect our commercial
prospects.
Before obtaining marketing approval from regulatory authorities for
the sale of our product candidates, we must conduct extensive
clinical studies to demonstrate the safety, purity, potency and
efficacy of the product candidates in humans. Before we can
initiate clinical trials for our preclinical product candidates, we
must submit the results of preclinical studies to the FDA or
comparable foreign regulatory authorities along with other
information, including information about product candidate
chemistry, manufacturing and controls and our proposed clinical
trial protocol, as part of an IND application or similar regulatory
submission. The FDA or comparable foreign regulatory authorities
may require us to conduct additional preclinical studies for any
product candidate before it allows us to initiate clinical trials
under any IND or similar regulatory submission, which may lead to
delays and increase the costs of our preclinical development
programs. Moreover, even if these trials begin, issues may arise
that could cause regulatory authorities to suspend or terminate
such clinical trials. Any delays in the commencement or completion
of our ongoing and planned clinical trials for our current and any
future product candidate could significantly affect our product
development timelines and product development costs.
We do not know whether our planned trials will begin on time or if
our ongoing or future clinical trials will be completed on
schedule, if at all. The commencement, data readouts and completion
of clinical trials can be delayed for a number of reasons,
including delays related to:
•
inability to generate sufficient preclinical, toxicology, or other
in vivo or in vitro data to support the initiation or continuation
of clinical trial;
•
obtaining regulatory authorizations to commence a trial or reaching
a consensus with regulatory authorities on trial
design;
81
•
the FDA or comparable foreign regulatory authorities disagreeing as
to the design or implementation of our clinical
studies;
•
any failure or delay in reaching an agreement with CROs and
clinical trial sites, the terms of which can be subject to
extensive negotiation and may vary significantly among different
CROs and trial sites;
•
delays in identifying, recruiting and training suitable clinical
investigators;
•
obtaining approval from one or more IRBs at clinical trial
sites;
•
IRBs refusing to approve, suspending or terminating the trial at an
investigational site, precluding enrollment of additional subjects,
or withdrawing their approval of the trial;
•
changes to the clinical trial protocol;
•
clinical sites deviating from the trial protocol or dropping out of
a trial;
•
failure by us or our CROs to perform in accordance with GCP
requirements or applicable regulatory guidelines in other
countries;
•
manufacturing sufficient quantities of product candidates or
obtaining sufficient quantities of combination therapies for use in
clinical trials;
•
subjects failing to enroll or remain in our trials at the rate we
expect, or failing to return for post-treatment follow-up,
including subjects failing to remain in our trials due to movement
restrictions, heath reasons or otherwise resulting from the
COVID-19 pandemic;
•
patients choosing alternative treatments for the indications for
which we are developing our product candidates, or participating in
competing clinical trials;
•
lack of adequate funding to continue the clinical trials or costs
being greater than we anticipate;
•
subjects experiencing severe or unexpected drug-related adverse
effects;
•
occurrence of serious adverse events in trials of the same class of
agents conducted by other companies;
•
selection of clinical endpoints that require prolonged periods of
clinical observation or analysis of the resulting
data;
•
transfer of manufacturing processes to larger-scale facilities
operated by a contract manufacturing organization (CMO), delays or
failure by our CMOs or us to make any necessary changes to such
manufacturing process, or failure of our CMOs to produce clinical
trial materials in accordance with cGMP regulations or other
applicable requirements; and
•
third parties being unwilling or unable to satisfy their
contractual obligations to us in a timely manner.
In addition, disruptions caused by the COVID-19 pandemic may
increase the likelihood that we encounter such difficulties or
delays in initiating, enrolling, conducting or completing our
planned and ongoing clinical trials.
Clinical trials must be conducted in accordance with the FDA and
other applicable regulatory authorities’ legal requirements,
regulations or guidelines, and are subject to oversight by these
governmental agencies and Ethics Committees or IRBs at the medical
institutions where the clinical trials are conducted. We could also
encounter delays if a clinical trial is suspended or terminated by
us, by the IRBs of the institutions in which such trials are being
conducted, by a Data Safety Monitoring Board for such trial or by
the FDA or comparable foreign regulatory authorities. Such
authorities may impose such a suspension or termination due to a
number of factors, including failure to conduct the clinical trial
in accordance with regulatory requirements or our clinical
protocols, inspection of the clinical trial operations or trial
site by the FDA or comparable foreign regulatory authorities
resulting in the imposition of a clinical hold, unforeseen safety
issues or adverse side effects, failure to demonstrate a benefit
from using a drug, changes in governmental regulations or
administrative actions or lack of adequate funding to continue the
clinical trial. In addition, changes in regulatory requirements and
policies may occur, and we may need to amend clinical trial
protocols to comply with these changes. Amendments may require us
to resubmit our clinical trial protocols to IRBs for reexamination,
which may impact the costs, timing or successful completion of a
clinical trial.
82
Further, our conduct of clinical trials in foreign countries
presents additional risks that may delay completion of our clinical
trials. These risks include the failure of enrolled patients in
foreign countries to adhere to clinical protocol as a result of
differences in healthcare services or cultural customs, managing
additional administrative burdens associated with foreign
regulatory schemes, as well as political and economic risks,
including war, relevant to such foreign countries.
Moreover, principal investigators for our clinical trials may serve
as scientific advisors or consultants to us from time to time and
receive compensation in connection with such services. Under
certain circumstances, we may be required to report some of these
relationships to the FDA or comparable foreign regulatory
authorities. The FDA or comparable foreign regulatory authority may
conclude that a financial relationship between us and a principal
investigator has created a conflict of interest or otherwise
affected interpretation of the study. The FDA or comparable foreign
regulatory authority may therefore question the integrity of the
data generated at the applicable clinical trial site and the
utility of the clinical trial itself may be jeopardized. This could
result in a delay in approval, or rejection, of our marketing
applications by the FDA or comparable foreign regulatory authority,
as the case may be, and may ultimately lead to the denial of
marketing approval of one or more of our product
candidates.
In addition, many of the factors that cause, or lead to, the
termination or suspension of, or a delay in the commencement or
completion of, clinical trials may also ultimately lead to the
denial of regulatory approval of a product candidate. We may make
formulation or manufacturing changes to our product candidates, in
which case we may need to conduct additional preclinical studies to
bridge our modified product candidates to earlier versions. Any
delays to our clinical trials that occur as a result could shorten
any period during which we may have the exclusive right to
commercialize our product candidates and our competitors may be
able to bring products to market before we do, and the commercial
viability of our product candidates could be significantly reduced.
Any of these occurrences may harm our business, financial condition
and prospects significantly.
We may find it difficult to enroll patients in our clinical trials.
If we encounter difficulties enrolling subjects in our clinical
trials, our clinical development activities could be delayed or
otherwise adversely affected.
We may not be able to initiate or continue clinical trials for our
product candidates if we are unable to identify and enroll a
sufficient number of eligible patients to participate in these
trials as required by the FDA or similar regulatory authorities
outside the United States. Subject enrollment, a significant factor
in the timeline of clinical trials, is affected by many factors
including the size and characteristics of the patient population,
the proximity of patients to clinical sites, the eligibility and
exclusion criteria for the trial, the design of the clinical trial,
the risk that enrolled patients will not complete a clinical trial,
our ability to recruit clinical trial investigators with the
appropriate competencies and experience, competing clinical trials
and clinicians’ and patients’ perceptions as to the potential
advantages and risks of the product candidate being studied in
relation to other available therapies, including any new products
that may be approved for the indications we are investigating as
well as any product candidates under development. We will be
required to identify and enroll a sufficient number of subjects for
each of our clinical trials. Potential subjects for any planned
clinical trials may not be adequately diagnosed or identified with
the diseases which we are targeting or may not meet the entry
criteria for such trials. In particular, because certain of our
product candidates are focused on patients with specific molecular
alterations within the RAS/MAPK pathway, our ability to enroll
eligible patients may be limited or may result in slower enrollment
than we anticipate. We also may encounter difficulties in
identifying and enrolling patients with a stage of disease
appropriate for our planned clinical trials and monitoring such
patients adequately during and after treatment. Additionally, other
pharmaceutical companies targeting these same types of cancer are
recruiting clinical trial patients from these patient populations,
which may make it more difficult to fully enroll our clinical
trials. We may not be able to initiate or continue clinical trials
if we are unable to locate a sufficient number of eligible subjects
to participate in the clinical trials required by the FDA or
comparable foreign regulatory authorities. In addition, the process
of finding and diagnosing patients may prove costly. The timing of
our clinical trials depends, in part, on the speed at which we can
recruit patients to participate in our trials, as well as
completion of required follow-up periods. The eligibility criteria
of our clinical trials, once established, will further limit the
pool of available trial participants. If patients are unwilling to
participate in our trials for any reason, including the existence
of concurrent clinical trials for similar patient populations, the
availability of approved therapies or as a result of the COVID-19
pandemic, or we otherwise have difficulty enrolling a sufficient
number of patients, the timeline for recruiting subjects,
conducting studies and obtaining regulatory approval of our product
candidates may be delayed. Additionally, because our clinical
trials are in patients with relapsed/refractory cancer, the
patients are typically in the late stages of their disease and may
experience disease progression independent from our product
candidates, making them unevaluable for purposes of the clinical
trial and requiring additional patient enrollment. Our inability to
enroll a sufficient number of subjects for any of our future
clinical trials would result in significant delays or may require
us to abandon one or more clinical trials altogether. In addition,
we expect to rely on CROs and clinical trial sites to ensure proper
and timely conduct of our future clinical trials and, while we have
entered into agreements governing their services, we have limited
influence over their actual performance. We cannot assure you that
our
83
assumptions used in determining expected clinical trial timelines
are correct or that we will not experience delays in enrollment,
which would result in the delay of completion of such trials beyond
our expected timelines.
Use of our product candidates could be associated with side
effects, adverse events or other properties or safety risks, which
could delay or preclude approval, cause us to suspend or
discontinue clinical trials, abandon a product candidate, limit the
commercial profile of an approved label or result in other
significant negative consequences that could severely harm our
business, prospects, operating results and financial
condition.
As is the case with oncology drugs generally, it is likely that
there may be side effects and adverse events associated with our
product candidates’ use. Results of our clinical trials could
reveal a high and unacceptable severity and prevalence of side
effects or unexpected characteristics. Undesirable side effects
caused by our product candidates when used alone or in combination
with other approved or investigational drugs or biologics could
cause us or regulatory authorities to interrupt, delay or halt
clinical trials and could result in a more restrictive label, or
lead to the delay or denial of regulatory approval by the FDA or
comparable foreign regulatory authorities. The drug-related side
effects could affect patient recruitment or the ability of enrolled
patients to complete the trial or result in potential product
liability claims. Any of these occurrences may harm our business,
financial condition and prospects significantly.
Moreover, if our product candidates are associated with undesirable
side effects in clinical trials or have characteristics that are
unexpected, we may elect to abandon their development or limit
their development to more narrow uses or subpopulations in which
the undesirable side effects or other characteristics are less
prevalent, less severe or more acceptable from a risk-benefit
perspective, which may limit the commercial expectations for the
product candidate if approved. For example, ophthalmic toxicities
have been observed during treatment with MEK-targeted agents and
also occur with ERK inhibitors, and reversible retinopathy is a
well-known MEK/ERK class effect. Skin toxicities have also been
noted as a class effect of inhibitors of RAF, MEK, and ERK. Both
skin and ophthalmic treatment-related adverse events were observed
in the completed Phase 1 trial of ERAS-007, consistent with these
class effects. Gastrointestinal toxicities are associated with the
use of MEK/ERK inhibitors and SHP2 inhibitors, whereas
hematological toxicities are more commonly associated with SHP2
inhibitors. Furthermore, skin and gastrointestinal side effects
represent overlapping toxicities of ERK inhibitors and SHP2
inhibitors with EGFR inhibitors and BRAF inhibitors. Therefore,
unacceptable enhancement of certain toxicities may be seen when our
product candidates are combined with standard of care therapies, or
when they are used as single agents. We may also be required to
modify our development and clinical trial plans based on findings
in our ongoing clinical trials. Many compounds that initially
showed promise in early-stage testing for treating cancer have
later been found to cause side effects that prevented further
development of the compound. In addition, regulatory authorities
may draw different conclusions or require additional testing to
confirm these determinations.
It is possible that as we test our product candidates in larger,
longer and more extensive clinical trials, including with different
dosing regimens, or as the use of these product candidates becomes
more widespread if they receive regulatory approval, illnesses,
injuries, discomforts and other adverse events that were observed
in earlier trials, as well as conditions that did not occur or went
undetected in previous trials, may be reported by subjects. If such
side effects become known later in development or upon approval, if
any, such findings may harm our business, financial condition and
prospects significantly.
In addition, we plan to study our product candidates in combination
with other therapies, including those that are also known to act on
the RAS/MAPK pathway, which may exacerbate adverse events
associated with such product candidates. Patients treated with our
product candidates may also be undergoing surgical, radiation and
chemotherapy treatments, which can cause side effects or adverse
events that are unrelated to our product candidates but may still
impact the success of our clinical trials. The inclusion of
critically ill patients in our clinical trials may result in deaths
or other adverse medical events due to other therapies or
medications that such patients may be using or due to the gravity
of such patients’ illnesses. For example, it is expected that some
of the patients enrolled in our clinical trials will die or
experience major clinical events either during the course of our
clinical trials or after participating in such trials, which has
occurred in the past.
In addition, if one or more of our product candidates receives
marketing approval, and we or others later identify undesirable
side effects caused by such product, a number of potentially
significant negative consequences could result,
including:
•
regulatory authorities may withdraw, suspend or limit approvals of
such product, or seek an injunction against its manufacture or
distribution;
•
we may be required to recall a product or change the way such
product is administered to patients;
84
•
regulatory authorities may require additional warnings on the
label, such as a “black box” warning or a
contraindication;
•
we may be required to implement a REMS or create a medication guide
outlining the risks of such side effects for distribution to
patients;
•
we may be required to change the way a product is distributed or
administered, conduct additional clinical trials or change the
labeling of a product or be required to conduct additional
post-marketing studies or surveillance;
•
we could be sued and held liable for harm caused to
patients;
•
sales of the product may decrease significantly or the product
could become less competitive; and
•
our reputation may suffer.
Any of these events could prevent us from achieving or maintaining
market acceptance of the particular product candidate, if approved,
and could significantly harm our business, results of operations
and prospects.
As an organization, we have never completed any clinical trials and
may be unable to do so for any of our product
candidates.
We are early in our development efforts for our product candidates,
have never completed any clinical trials and we will need to
successfully complete our Phase 1 clinical trials and later-stage
and pivotal clinical trials in order to obtain FDA or comparable
foreign regulatory approval to market our product candidates.
Carrying out later-stage clinical trials and the submission of a
successful NDA or BLA or similar regulatory submissions to
comparable foreign regulatory authorities is a complicated process.
A Phase 1 clinical trial for ERAS-007 was completed prior to our
acquisition of this product candidate, and we are currently
conducting our first Phase 1 clinical trials for ERAS-601 and
ERAS-801, and multiple Phase 1b/2 trials for ERAS-007. We are only
beginning to conduct clinical trials for our product candidates,
and we have limited experience as a company in preparing,
submitting and prosecuting regulatory filings and have not
previously submitted an NDA, BLA or other comparable foreign
regulatory submission for any product candidate. We are also
conducting and plan to conduct a number of clinical trials for
multiple product candidates in parallel over the next several
years, which may be a difficult process to manage with our limited
resources and which may divert the attention of management. In
addition, we have had limited interactions with the FDA or other
comparable foreign regulatory authorities, and cannot be certain
how many additional clinical trials of our product candidates will
be required or how such trials should be designed. Consequently, we
may be unable to successfully and efficiently execute and complete
necessary clinical trials in a way that leads to regulatory
submission and approval of any of our product candidates. We may
require more time and incur greater costs than our competitors and
may not succeed in obtaining regulatory approvals of product
candidates that we develop. Failure to commence or complete, or
delays in, our planned clinical trials could prevent us from or
delay us in submitting marketing applications, including NDAs and
BLAs, and commercializing our product candidates.
We intend to develop our product candidates in combination with
other therapies, which exposes us to additional risks.
We intend to develop our current and any future product candidates
for use in combination with one or more currently approved cancer
therapies. Even if any product candidate we develop was to receive
marketing approval or be commercialized for use in combination with
other existing therapies, we would continue to bear the risks that
the FDA or similar foreign regulatory authorities could revoke
approval of the therapy used in combination with our product
candidate or that safety, efficacy, manufacturing or supply issues
could arise with these existing therapies. Combination therapies
are commonly used for the treatment of cancer, and we would be
subject to similar risks if we develop any of our product
candidates for use in combination with other drugs or biologics or
for indications other than cancer. Developing combination therapies
using approved therapeutics, as we plan to do for our product
candidates, also exposes us to additional clinical risks, such as
the requirement that we demonstrate the safety and efficacy of each
active component of any combination regimen we may
develop.
In addition, we are also evaluating the combination of ERAS-007 and
ERAS-601 with each other, and may also evaluate our product
candidates in combination with one or more other cancer therapies
that have not yet been approved for marketing by the FDA or similar
foreign regulatory authorities. We may not be able to market and
sell any product candidate we develop in combination with any such
unapproved cancer therapies that do not ultimately obtain marketing
approval.
85
If the FDA or similar foreign regulatory authorities do not approve
these other combination agents or revoke their approval of, or if
safety, efficacy, manufacturing, or supply issues arise with the
drugs or biologics we choose to evaluate in combination with our
product candidates, we may be unable to obtain approval of or
market our product candidates for combination therapy
regimens.
Additionally, if the third-party providers of therapies or
therapies in development used in combination with our product
candidates are unable to produce sufficient quantities for clinical
trials or for commercialization of our product candidates, or if
the cost of combination therapies are prohibitive, our development
and commercialization efforts would be impaired, which would have
an adverse effect on our business, financial condition, results of
operations and growth prospects.
Because we have a number of product candidates and development
programs in our pipeline, we may expend our limited resources to
pursue a particular product candidate and fail to capitalize on
product candidates or indications that may be more profitable or
for which there is a greater likelihood of success.
Because we have limited financial and managerial resources, we
focus on specific product candidates, development programs and
indications. We are also conducting and plan to conduct several
clinical trials for multiple product candidates in parallel over
the next several years, which may make our decision as to which
product candidates to focus on more difficult. As a result, we may
forgo or delay pursuit of opportunities with other product
candidates that could have had greater commercial potential. Our
resource allocation decisions may cause us to fail to capitalize on
viable commercial products or profitable market opportunities. Our
spending on current and future research and development programs
and product candidates for specific indications may not yield any
commercially viable product candidates. If we do not accurately
evaluate the commercial potential or target market for a particular
product candidate, we may relinquish valuable rights to that
product candidate through collaborations, licenses and other
similar arrangements in cases in which it would have been more
advantageous for us to retain sole development and
commercialization rights to such product candidate.
Additionally, we may pursue additional in-licenses or acquisitions
of development-stage assets or programs, which entails additional
risk to us. Identifying, selecting and acquiring promising product
candidates requires substantial technical, financial and human
resources expertise. Efforts to do so may not result in the actual
acquisition or license of a particular product candidate,
potentially resulting in a diversion of our management’s time and
the expenditure of our resources with no resulting benefit. For
example, if we are unable to identify programs that ultimately
result in approved products, we may spend material amounts of our
capital and other resources evaluating, acquiring and developing
products that ultimately do not provide a return on our
investment.
We may not be able to obtain or maintain orphan designations for
any of our product candidates, and we may be unable to maintain the
benefits associated with orphan drug designation, including the
potential for market exclusivity.
We may seek orphan designation for some of our product candidates;
however, we may never receive such designations. Under the Orphan
Drug Act of 1983, the FDA may designate a product as an orphan
product candidate if it is intended to treat a rare disease or
condition, which is generally defined as a patient population of
fewer than 200,000 individuals in the United States, or a patient
population of greater than 200,000 individuals in the United
States, but for which there is no reasonable expectation that the
cost of developing the drug or biologic will be recovered from
sales in the United States. Orphan drug designation must be
requested before submitting an NDA or BLA.
In the United States, orphan designation entitles a party to
financial incentives such as opportunities for grant funding toward
clinical trial costs, tax advantages and user-fee waivers. In
addition, if a product candidate that has orphan designation
subsequently receives the first FDA approval for the disease for
which it has such designation, the product is entitled to orphan
drug exclusivity, which means that the FDA may not approve any
other applications, including an NDA or BLA, to market the same
product for the same indication for seven years, except in limited
circumstances, such as a showing of clinical superiority to the
product with orphan drug exclusivity or where the manufacturer is
unable to assure sufficient product quantity.
Even if we obtain orphan drug exclusivity for a product, such
exclusivity may not effectively protect the product from
competition because different drugs and biologics can be approved
for the same condition. Even after an orphan drug or biologic is
approved, the FDA or comparable foreign regulatory authority can
subsequently approve the same drug or biologic for the same
condition if such regulatory authority concludes that the later
drug or biologic is clinically superior because it is shown to be
safer, more effective or makes a major contribution to patient
care. Orphan drug exclusivity may also be lost if the FDA later
determines that the initial request for designation was materially
defective, or if the sponsor
86
seeks approval for an indication broader than the designated
indication. In addition, orphan drug exclusivity does not prevent
the FDA from approving competing drugs or biologics for the same or
similar indication containing a different active ingredient. In
addition, if a subsequent drug or biologic is approved for
marketing for the same or a similar indication as any of our
product candidates that receive marketing approval, we may face
increased competition and lose market share regardless of orphan
drug exclusivity. Orphan designation neither shortens the
development time or regulatory review time of a drug nor gives the
drug any advantage in the regulatory review or approval process.
Where applicable, we also may seek comparable designations for our
product candidates in other jurisdictions, which may have differing
requirements and would also include risks of not being granted
and/or not being effective in protecting the product from
competition.
We are currently conducting and may in the future conduct certain
of our clinical trials for our product candidates outside of the
United States. However, the FDA and other foreign equivalents may
not accept data from such trials, in which case our development
plans will be delayed, which could materially harm our
business.
We are currently conducting and may in the future conduct one or
more of our clinical trials for our product candidates outside the
United States. The acceptance of data from clinical trials
conducted outside the United States or another jurisdiction by the
FDA or comparable foreign regulatory authority may be subject to
certain conditions or may not be accepted at all. In cases where
data from foreign clinical trials are intended to serve as the sole
basis for marketing approval in the United States, the FDA will
generally not approve the application on the basis of foreign data
alone unless (i) the data are applicable to the US population and
US medical practice; (ii) the trials were performed by clinical
investigators of recognized competence and pursuant to GCP
regulations; and (iii) the data may be considered valid without the
need for an on-site inspection by the FDA, or if the FDA considers
such inspection to be necessary, the FDA is able to validate the
data through an on-site inspection or other appropriate means. In
addition, even where the foreign clinical trial data are not
intended to serve as the sole basis for approval, the FDA will not
accept the data as support for an application for marketing
approval unless the trial is well-designed and well-conducted in
accordance with GCP requirements and the FDA is able to validate
the data from the trial through an onsite inspection if deemed
necessary. Many foreign regulatory authorities have similar
approval requirements. In addition, such foreign trials would be
subject to the applicable local laws of the foreign jurisdictions
where the trials are conducted. There can be no assurance that the
FDA or any comparable foreign regulatory authority will accept data
from trials conducted outside of the United States or the
applicable jurisdiction. If the FDA or any comparable foreign
regulatory authority does not accept such data, it would result in
the need for additional trials, which could be costly and
time-consuming, and which may result in current or future product
candidates that we may develop not receiving approval for
commercialization in the applicable jurisdiction.
Conducting clinical trials outside the United States also exposes
us to additional risks, including risks associated with:
•
additional foreign regulatory requirements;
•
foreign exchange fluctuations;
•
compliance with foreign manufacturing, customs, shipment and
storage requirements;
•
inconsistent standards for reporting and evaluating clinical data
and adverse events;
•
cultural differences in medical practice and clinical research;
and
•
diminished protection of intellectual property in some
countries.
Interim, topline and preliminary data from our clinical trials and
preclinical studies that we announce or publish from time to time
may change as more patient data become available and are subject to
audit and verification procedures that could result in material
changes in the final data.
From time to time, we may publicly disclose interim, top-line, or
preliminary data from our clinical trials and preclinical studies,
which is based on a preliminary analysis of then-available data,
and the results and related findings and conclusions are subject to
change following a full analyses of all data related to the
particular trial. We also make assumptions, estimations,
calculations and conclusions as part of our analyses of data, and
we may not have received or had the opportunity to fully and
carefully evaluate all data. As a result, the interim, top-line, or
preliminary results that we report may differ from future results
of the same trials, or different conclusions or considerations may
qualify such results, once additional data have been received and
fully evaluated. Top-line and preliminary data also remain subject
to audit and verification procedures that may result in the final
data being materially different from the preliminary data we
previously published. As a result, top-line and preliminary data
should be viewed with caution until the final data are available.
We may also disclose interim data from our clinical
trials.
87
Interim data from clinical trials that we may complete are also
subject to the risk that one or more of the clinical outcomes may
materially change as patient enrollment continues and more patient
data become available. Adverse differences between interim,
top-line, or preliminary data and final data could significantly
harm our business prospects.
Further, others, including regulatory agencies, may not accept or
agree with our assumptions, estimates, calculations, conclusions or
analyses or may interpret or weigh the importance of data
differently, which could impact the value of the particular
program, the approvability or commercialization of the particular
product candidate or product and our business in general. In
addition, the information we choose to publicly disclose regarding
a particular study or clinical trial is based on what is typically
extensive information, and you or others may not agree with what we
determine is the material or otherwise appropriate information to
include in our disclosure, and any information we determine not to
disclose may ultimately be deemed significant with respect to
future decisions, conclusions, views, activities or otherwise
regarding a particular drug, product candidate or our business. If
the interim, top-line, or preliminary data that we report differ
from actual results, or if others, including regulatory
authorities, disagree with the conclusions reached, our ability to
obtain approval for and commercialize our product candidates, our
business, operating results, prospects or financial condition may
be harmed.
We may attempt to secure approval from the FDA or comparable
foreign regulatory authorities through the use of accelerated
approval pathways. If we are unable to obtain such approval, we may
be required to conduct additional clinical trials beyond those that
we contemplate, which could increase the expense of obtaining, and
delay the receipt of, necessary marketing approvals. Even if we
receive accelerated approval from the FDA (or a similar expedited
approval mechanism from a comparable foreign regulatory authority,
such as conditional marketing authorization in the EU), if our
confirmatory trials do not verify clinical benefit, or if we do not
comply with rigorous post-marketing requirements, the FDA (or
comparable foreign regulatory authority) may seek to withdraw
accelerated approval.
We may in the future seek an expedited approval for one or more of
our product candidates. Under the accelerated approval program, the
FDA or comparable foreign regulatory authority may grant
accelerated or conditional approval to a product candidate designed
to treat a serious or life-threatening condition that provides
meaningful therapeutic benefit over available therapies upon a
determination that the product candidate has an effect on a
surrogate endpoint or intermediate clinical endpoint that is
reasonably likely to predict clinical benefit. The health
authorities assess the definition of available therapy at the time
of regulatory decision-making; it is possible that the standard of
care may evolve during a drug’s development, for example, due to a
new therapy receiving full approval for that indication. In this
situation, an expedited approval, such as accelerated or
conditional approval, would be assessed for benefit over the newly
approved treatment which could require a better benefit/risk ratio
for approval, compared to the previous standard of care. The FDA
considers a clinical benefit to be a positive therapeutic effect
that is clinically meaningful in the context of a given disease,
such as irreversible morbidity or mortality. For the purposes of
accelerated approval, a surrogate endpoint is a marker, such as a
laboratory measurement, radiographic image, physical sign, or other
measure that is thought to predict clinical benefit, but is not
itself a measure of clinical benefit. An intermediate clinical
endpoint is a clinical endpoint that can be measured earlier than
an effect on irreversible morbidity or mortality that is reasonably
likely to predict an effect on irreversible morbidity or mortality
or other clinical benefit. The accelerated approval pathway may be
used in cases in which the advantage of a new drug or biologic over
available therapy may not be a direct therapeutic advantage, but is
a clinically important improvement from a patient and public health
perspective. If granted, accelerated approval and conditional
approval are contingent on the sponsor’s agreement to conduct, in a
diligent manner, additional post- approval confirmatory studies to
verify and describe the drug’s clinical benefit. If such
post-approval studies fail to confirm the drug or biologic’s
clinical benefit or are not completed in a timely manner, the FDA
or comparable foreign regulatory authority may withdraw its
approval of the drug or biologic.
Prior to seeking approval for any of our product candidates, we
intend to seek feedback from the applicable health authorities,
such as the FDA and will otherwise evaluate our ability to seek and
receive accelerated or conditional approval. There can be no
assurance that after our evaluation of the feedback and other
factors we will decide to pursue or submit an NDA, BLA, or
comparable foreign marketing application for accelerated approval
or any other form of expedited development, review or approval
mechanism.
Similarly, there can be no assurance that after subsequent health
authority feedback we will continue to pursue or apply for
accelerated approval or any other form of expedited development,
review or approval mechanism, even if we initially decide to do so.
Furthermore, if we decide to submit an application for accelerated
or conditional approval, or receive an expedited regulatory
designation (e.g., breakthrough therapy designation) for our
product candidates, there can be no assurance that such submission
or application will be accepted or that any expedited development,
review or approval will be granted on a timely basis, or at all.
The FDA or other comparable foreign regulatory authority could also
require us to
88
conduct further studies prior to considering our application or
granting approval of any type. A failure to obtain accelerated
approval or any other form of expedited development, review or
approval for our product candidate would result in a longer time
period to commercialization of such product candidate, if any,
could increase the cost of development of such product candidate
and could harm our competitive position in the
marketplace.
Disruptions at the FDA and other government agencies caused by
funding shortages or global health concerns could hinder their
ability to hire, retain or deploy key leadership and other
personnel, or otherwise prevent new or modified products from being
developed, approved or commercialized in a timely manner or at all,
which could negatively impact our business.
The ability of the FDA to review and approve new products can be
affected by a variety of factors, including government budget and
funding levels, statutory, regulatory and policy changes, the FDA’s
ability to hire and retain key personnel and accept the payment of
user fees, and other events that may otherwise affect the FDA’s
ability to perform routine functions. Average review times at the
FDA have fluctuated in recent years. In addition, government
funding of other government agencies that fund research and
development activities is subject to the political process, which
is inherently fluid and unpredictable. Disruptions at the FDA and
other agencies may also slow the time necessary for new drugs and
biologics or modifications to approved drugs and biologics to be
reviewed and/or approved by necessary government agencies, which
would adversely affect our business. For example, over the last
several years, the US government has shut down several times and
certain regulatory agencies, such as the FDA, have had to furlough
critical FDA employees and stop critical activities.
Separately, in response to the COVID-19 pandemic, in March 2020,
the FDA announced its intention to postpone most inspections of
foreign manufacturing facilities, and on March 18, 2020, the FDA
temporarily postponed routine surveillance inspections of domestic
manufacturing facilities. Subsequently, in July 2020, the FDA
resumed certain on-site inspections of domestic manufacturing
facilities subject to a risk-based prioritization system. The FDA
utilized this risk-based assessment system to assist in determining
when and where it was safest to conduct prioritized domestic
inspections. Additionally, on April 15, 2021, the FDA issued a
guidance document in which the FDA described its plans to conduct
voluntary remote interactive evaluations of certain drug
manufacturing facilities and clinical research sites, among other
facilities. According to the guidance, the FDA may request such
remote interactive evaluations where the FDA determines that remote
evaluation would be appropriate based on mission needs and travel
limitations. In May 2021, the FDA outlined a detailed plan to move
toward a more consistent state of inspectional operations, and in
July 2021, the FDA resumed standard inspectional operations of
domestic facilities and was continuing to maintain this level of
operation as of September 2021. More recently, the FDA has
continued to monitor and implement changes to its inspectional
activities to ensure the safety of its employees and those of the
firms it regulates as it adapts to the evolving COVID-19 pandemic.
Regulatory authorities outside the United States may adopt similar
restrictions or other policy measures in response to the COVID-19
pandemic. If a prolonged government shutdown occurs, or if global
health concerns continue to prevent the FDA or other regulatory
authorities from conducting their regular inspections, reviews or
other regulatory activities, it could significantly impact the
ability of the FDA or other regulatory authorities to timely review
and process our regulatory submissions, which could have a material
adverse effect on our business.
If we are required by the FDA
or comparable foreign regulatory authority
to obtain approval of a companion diagnostic test in connection
with approval of any of our product candidates, and we do not
obtain or face delays in obtaining FDA or foreign approval of a
diagnostic device, we may not be able to commercialize such product
candidate and our ability to generate revenue will be materially
impaired.
If safe and effective use of any of our product candidates depends
on an
in vitro
diagnostic that is not otherwise commercially available, then the
FDA generally may require approval or clearance of that diagnostic,
known as a companion diagnostic, at the same time that the FDA
approves our product candidates, if at all. According to FDA
guidance, if the FDA determines that a companion diagnostic device
is essential to the safe and effective use of a novel therapeutic
product or indication, the FDA generally will not approve the
therapeutic product or new therapeutic product indication if the
companion diagnostic is not also approved or cleared for that
indication. If a satisfactory companion diagnostic is not
commercially available, we may be required to develop or obtain one
that would be subject to regulatory approval requirements. The
process of obtaining or creating such diagnostics is time-consuming
and costly.
Companion diagnostics are developed in conjunction with clinical
programs for the associated product and are subject to regulation
as medical devices by the FDA and comparable regulatory
authorities, and, to date, the FDA has generally required premarket
approval of companion diagnostics for cancer therapies. The
approval of a companion diagnostic as part of the therapeutic
product’s labeling limits the use of the therapeutic product to
only those patients who express the specific genetic alteration
that the companion diagnostic was developed to detect.
89
If the FDA or a comparable regulatory authority requires approval
of a companion diagnostic for any of our product candidates,
whether before, simultaneously with, or after such candidate
obtains marketing approval, if ever, we, and/or future
collaborators, may encounter difficulties in developing and
obtaining approval for such companion diagnostic. Any delay or
failure by us or third-party collaborators to develop or obtain
regulatory approval of a companion diagnostic could delay or
prevent approval or continued marketing of such product candidate.
We may also experience delays in developing a sustainable,
reproducible and scalable manufacturing process for the companion
diagnostic or in transferring that process to commercial partners
or negotiating insurance reimbursement plans, all of which may
prevent us from completing our clinical trials or commercializing
our product candidate, if approved, on a timely or profitable
basis, if at all.
Risks related to our reliance on third parties
We rely on third parties to conduct our clinical trials and
preclinical studies. If these third parties do not successfully
carry out their contractual duties, comply with applicable
regulatory requirements or meet expected deadlines, our development
programs and our ability to seek or obtain regulatory approval for
or commercialize our product candidates may be delayed.
We are dependent on third parties to conduct our clinical trials
and preclinical studies. Specifically, we have used and relied on,
and intend to continue to use and rely on, medical institutions,
clinical investigators, CROs and consultants to conduct our
preclinical studies and clinical trials in accordance with our
clinical protocols and regulatory requirements. These CROs,
investigators and other third parties play a significant role in
the conduct and timing of these trials and subsequent collection
and analysis of data. While we have and will have agreements
governing the activities of our third-party contractors, we have
limited influence over their actual performance. Nevertheless, we
are responsible for ensuring that each of our clinical trials is
conducted in accordance with the applicable protocol and legal,
regulatory and scientific standards, and our reliance on our CROs
and other third parties does not relieve us of our regulatory
responsibilities. We and our CROs are required to comply with GCP
requirements, which are regulations and guidelines enforced by the
FDA and comparable foreign regulatory authorities for all of our
product candidates in clinical development. Regulatory authorities
enforce these GCPs through periodic inspections of trial sponsors,
principal investigators and trial sites. If we or any of our CROs
or trial sites fail to comply with applicable GCPs, the clinical
data generated in our clinical trials may be deemed unreliable, and
the FDA or comparable foreign regulatory authorities may require us
to perform additional clinical trials before approving our
marketing applications. In addition, our clinical trials must be
conducted with product candidates and products produced under cGMP
regulations. Our failure to comply with these regulations may
require us to repeat clinical trials, which would delay the
regulatory approval process.
There is no guarantee that any of our CROs, investigators or other
third parties will devote adequate time and resources to such
trials or studies or perform as contractually required. If any of
these third parties fail to meet expected deadlines, adhere to our
clinical protocols or meet regulatory requirements, or otherwise
performs in a substandard manner, our clinical trials may be
extended, delayed or terminated. In addition, many of the third
parties with whom we contract may also have relationships with
other commercial entities, including our competitors, for whom they
may also be conducting clinical trials or other development
activities that could harm our competitive position. In addition,
principal investigators for our clinical trials are expected to
serve as scientific advisors or consultants to us from time to time
and may receive cash or equity compensation in connection with such
services. If these relationships and any related compensation
result in perceived or actual conflicts of interest, or the FDA
concludes that the financial relationship may have affected the
interpretation of the study, the integrity of the data generated at
the applicable clinical trial site may be questioned and the
utility of the clinical trial itself may be jeopardized, which
could result in the delay or rejection by the FDA of any NDA or BLA
we submit. Any such delay or rejection could prevent us from
commercializing our product candidates.
Our CROs have the right to terminate their agreements with us in
the event of an uncured material breach. In addition, some of our
CROs have an ability to terminate their respective agreements with
us if it can be reasonably demonstrated that the safety of the
subjects participating in our clinical trials warrants such
termination, if we make a general assignment for the benefit of our
creditors or if we are liquidated. If any of our relationships with
these third parties terminate, we may not be able to enter into
arrangements with alternative third parties on commercially
reasonable terms or at all. Switching or adding additional CROs,
investigators and other third parties involves additional cost and
requires our management’s time and focus. In addition, there is a
natural transition period when a new CRO commences work. As a
result, delays occur, which can materially impact our ability to
meet our desired clinical development timelines. Though we
carefully manage our relationships with our CROs, investigators and
other third parties, there can be no assurance that we will not
encounter challenges or delays in the future or that these delays
or challenges will not have a material adverse impact on our
business, financial condition and prospects.
90
We rely on third parties for the manufacture of our product
candidates for clinical and preclinical development and expect to
continue to do so for the foreseeable future. This reliance on
third parties increases the risk that we will not have sufficient
quantities of our product candidates or products or such quantities
at an acceptable cost, which could delay, prevent or impair our
development or commercialization efforts.
We do not own or operate manufacturing facilities and have no plans
to develop our own clinical or commercial-scale manufacturing
capabilities. We rely, and expect to continue to rely, on third
parties for the manufacture of our product candidates and related
raw materials for clinical and preclinical development, as well as
for commercial manufacture if any of our product candidates receive
marketing approval. The facilities used by third-party
manufacturers to manufacture our product candidates must be
approved by the FDA and any comparable foreign regulatory authority
pursuant to inspections that will be conducted after we submit an
NDA or BLA to the FDA or any comparable submission to a foreign
regulatory authority. We do not control the manufacturing process
of, and are completely dependent on, third-party manufacturers for
compliance with cGMP requirements for manufacture of product
candidates and products. If these third-party manufacturers cannot
successfully manufacture material that conforms to our
specifications and the strict regulatory requirements of the FDA or
any comparable foreign regulatory authority, they will not be able
to secure and/or maintain regulatory approval for their
manufacturing facilities. In addition, we have no control over the
ability of third-party manufacturers to maintain adequate quality
control, quality assurance and qualified personnel. If the FDA or
any comparable foreign regulatory authority does not approve these
facilities for the manufacture of our product candidates or if it
withdraws any such approval in the future, we may need to find
alternative manufacturing facilities, which would significantly
impact our ability to develop, obtain regulatory approval for or
market our product candidates, if approved. Our failure, or the
failure of our third-party manufacturers, to comply with applicable
regulations could result in sanctions being imposed on us,
including clinical holds, fines, injunctions, civil penalties,
delays, suspension or withdrawal of approvals, seizures or recalls
of product candidates or products, operating restrictions and
criminal prosecutions, any of which could significantly and
adversely affect supplies of our products.
Our or a third party’s failure to execute on our manufacturing
requirements on commercially reasonable terms and in compliance
with cGMP or other regulatory requirements could adversely affect
our business in a number of ways, including:
•
an inability to initiate clinical trials of our product candidates
under development;
•
delay in submitting regulatory applications, or receiving marketing
approvals, for our product candidates;
•
additional inspections by regulatory authorities of third-party
manufacturing facilities or our manufacturing
facilities;
•
requirements to cease development or to recall batches of our
product candidates; and
•
in the event of approval to market and commercialize a product, an
inability to meet commercial demands for such product.
In addition, we do not have any long-term commitments or supply
agreements with our third-party manufacturers. We may be unable to
establish any supply agreements with our third-party manufacturers
or to do so on acceptable terms, which increases the risk of timely
obtaining sufficient quantities of our product candidates or
products or such quantities at an acceptable cost. Even if we are
able to establish agreements with third-party manufacturers,
reliance on third-party manufacturers entails additional risks,
including:
•
failure of third-party manufacturers to comply with regulatory
requirements and maintain quality assurance;
•
breach of the manufacturing agreement by the third
party;
•
failure to manufacture our product according to our
specifications;
•
failure to manufacture our product according to our schedule or at
all;
•
misappropriation of our proprietary information, including our
trade secrets and know-how; and
•
termination or nonrenewal of the agreement by the third party at a
time that is costly or inconvenient for us.
Our product candidates and any products that we may develop may
compete with other product candidates and products for access to
manufacturing facilities. There are a limited number of
manufacturers that operate under cGMP regulations and that might be
capable of manufacturing for us, in particular due to the high
potency of our product candidates.
91
Any performance failure on the part of our existing or future
manufacturers could delay clinical development or marketing
approval, and any related remedial measures may be costly or
time-consuming to implement. We do not currently have arrangements
in place for redundant supply or a second source for all required
raw materials used in the manufacture of our product candidates. If
our existing or future third-party manufacturers cannot perform as
agreed, we may be required to replace such manufacturers and we may
be unable to replace them on a timely basis or at all. In
particular, any replacement of our manufacturers could require
significant effort and expertise because there may be a limited
number of qualified replacements. In some cases, the technical
skills or technology required to manufacture our product candidates
may be unique or proprietary to the original manufacturer and we
may have difficulty transferring such skills or technology to
another third-party and a feasible alternative may not exist. In
addition, certain of our product candidates and our own proprietary
methods have never been produced or implemented outside of our
company, and we may therefore experience delays to our development
programs if and when we attempt to establish new third-party
manufacturing arrangements for these product candidates or
methods.
Our current and anticipated future dependence upon others for the
manufacture of our product candidates or products may adversely
affect our future profit margins and our ability to commercialize
any products that receive marketing approval on a timely and
competitive basis.
Our reliance on third parties requires us to share our trade
secrets, which increases the possibility that a competitor will
discover them or that our trade secrets will be misappropriated or
disclosed.
Because we currently rely on third parties to manufacture our
product candidates and to perform quality testing, we must, at
times, share our proprietary technology and confidential
information, including trade secrets, with them. We seek to protect
our proprietary technology, in part, by entering into
confidentiality agreements, and, if applicable, material transfer
agreements, collaborative research agreements, consulting
agreements or other similar agreements with our collaborators,
advisors, employees and consultants prior to beginning research or
disclosing proprietary information. These agreements typically
limit the rights of the third parties to use or disclose our
confidential information. Despite the contractual provisions
employed when working with third parties, the need to share trade
secrets and other confidential information increases the risk that
such trade secrets become known by our competitors, are
intentionally or inadvertently incorporated into the technology of
others or are disclosed or used in violation of these agreements.
Given that our proprietary position is based, in part, on our
know-how and trade secrets and despite our efforts to protect our
trade secrets, a competitor’s discovery of our proprietary
technology and confidential information or other unauthorized use
or disclosure would impair our competitive position and may have a
material adverse effect on our business, financial condition,
results of operations and prospects.
We may seek to enter into collaborations, licenses and other
similar arrangements and may not be successful in doing so, and
even if we are, we may relinquish valuable rights and may not
realize the benefits of such relationships.
We may seek to enter into collaborations, joint ventures, licenses
and other similar arrangements for the development or
commercialization of our product candidates, due to capital costs
required to develop or commercialize the product candidate or
manufacturing constraints. For example, we are collaborating with
ELS on large molecule capabilities. Such collaborative discovery
efforts may not yield additional development or product candidates
for our pipeline. We may not be successful in our efforts to
establish or maintain such collaborations for our product
candidates because our research and development pipeline may be
insufficient, our product candidates may be deemed to be at too
early of a stage of development for collaborative effort or third
parties may not view our product candidates as having the requisite
potential to demonstrate safety and efficacy or significant
commercial opportunity. In addition, we face significant
competition in seeking appropriate strategic partners, and the
negotiation process can be time-consuming and complex. We may have
to relinquish valuable rights to our future revenue streams,
research programs or product candidates, or grant licenses on terms
that may not be favorable to us, as part of any such arrangement,
and such arrangements may restrict us from entering into additional
agreements with other potential collaborators. We cannot be certain
that, following a collaboration, license or strategic transaction,
we will achieve an economic benefit that justifies such
transaction.
Even if we are successful in our efforts to establish such
collaborations, the terms that we agree upon may not be favorable
to us, and we may not be able to maintain such collaborations if,
for example, the development or approval of a product candidate is
delayed, the safety of a product candidate is questioned or the
sales of an approved product candidate are
unsatisfactory.
92
In addition, any potential future collaborations may be terminable
by our strategic partners, and we may not be able to adequately
protect our rights under these agreements. Furthermore, strategic
partners may negotiate for certain rights to control decisions
regarding the development and commercialization of our product
candidates, if approved, and may not conduct those activities in
the same manner as we do. Any termination of collaborations we
enter into in the future, or any delay in entering into
collaborations related to our product candidates, could delay the
development and commercialization of our product candidates and
reduce their competitiveness if they reach the market, which could
have a material adverse effect on our business, financial condition
and results of operations.
Risks related to commercialization of our product
candidates
Even if we receive regulatory approval for any product candidate,
we will be subject to ongoing regulatory obligations and continued
regulatory review, which may result in significant additional
expense.
Any regulatory approvals that we may receive for our product
candidates will require the submission of reports to regulatory
authorities and surveillance to monitor the safety and efficacy of
the product, may contain significant limitations related to use
restrictions for specified age groups, warnings, precautions or
contraindications, and may include burdensome post-approval study
or risk management requirements. For example, the FDA may require a
REMS as a condition of approval of our product candidates, which
could include requirements for a medication guide, physician
communication plans or additional elements to ensure safe use, such
as restricted distribution methods, patient registries and other
risk minimization tools. In addition, if the FDA or a comparable
foreign regulatory authority approves our product candidates, the
manufacturing processes, labeling, packaging, distribution, adverse
event reporting, storage, advertising, promotion, import, export
and recordkeeping for our products will be subject to extensive and
ongoing regulatory requirements. These requirements include
submissions of safety and other post-marketing information and
reports, registration, as well as continued compliance with cGMP
and GCP requirements for any clinical trials that we conduct
post-approval. Manufacturers of approved products and their
facilities are subject to continual review and periodic,
unannounced inspections by the FDA and other regulatory authorities
for compliance with cGMP regulations and standards. Later discovery
of previously unknown problems with our products, including adverse
events of unanticipated severity or frequency, or with our
third-party manufacturers or manufacturing processes, or failure to
comply with regulatory requirements, may result in, among other
things:
•
restrictions on the marketing or manufacturing of our products,
withdrawal of the product from the market or voluntary or mandatory
product recalls;
•
restrictions on product distribution or use, or requirements to
conduct post-marketing studies or clinical trials;
•
fines, restitutions, disgorgement of profits or revenue, warning
letters, untitled letters or holds on clinical trials;
•
refusal by the FDA to approve pending applications or supplements
to approved applications filed by us or suspension or revocation of
approvals;
•
product seizure or detention, or refusal to permit the import or
export of our products; and
•
injunctions or the imposition of civil or criminal
penalties.
The occurrence of any event or penalty described above may inhibit
our ability to commercialize our product candidates and generate
revenue and could require us to expend significant time and
resources in response and could generate negative
publicity.
The FDA’s and other regulatory authorities’ policies may change and
additional government regulations may be enacted that could
prevent, limit or delay regulatory approval of our product
candidates. We also cannot predict the likelihood, nature or extent
of government regulation that may arise from future legislation or
administrative action, either in the United States or abroad. If we
are slow or unable to adapt to changes in existing requirements or
the adoption of new requirements or policies, or if we are not able
to maintain regulatory compliance, we may be subject to enforcement
action and we may not achieve or sustain profitability.
The FDA and other regulatory agencies actively enforce the laws and
regulations prohibiting the promotion of off-label uses.
If any of our product candidates are approved and we are found to
have improperly promoted off-label uses of those products, we may
become subject to significant liability. The FDA and other
regulatory agencies strictly regulate the promotional claims that
may be made about prescription products, such as our product
candidates, if approved. In particular, a product may not be
promoted for uses that are not approved by the FDA or such other
regulatory agencies as
93
reflected in the product’s approved labeling. If we receive
marketing approval for a product candidate, physicians may
nevertheless prescribe it to their patients in a manner that is
inconsistent with the approved label. If we are found to have
promoted such off-label uses, we may become subject to significant
liability. The US federal government has levied large civil and
criminal fines against companies for alleged improper promotion of
off-label use and has enjoined several companies from engaging in
off-label promotion. The government has also required companies to
enter into consent decrees or imposed permanent injunctions under
which specified promotional conduct is changed or curtailed. If we
cannot successfully manage the promotion of our product candidates,
if approved, we could become subject to significant liability,
which would materially adversely affect our business and financial
condition.
Our product candidates for which we intend to seek approval as
biologic products may face competition sooner than
anticipated.
The ACA includes the BPCIA, which created an abbreviated approval
pathway for biological products that are biosimilar to or
interchangeable with an FDA-licensed reference biological product.
Under the BPCIA, an application for a highly similar or
“biosimilar” product may not be submitted to the FDA until four
years following the date that the reference product was first
approved by the FDA. In addition, the approval of a biosimilar
product may not be made effective by the FDA until 12 years from
the date on which the reference product was first approved. During
this 12-year period of exclusivity, another company may still
market a competing version of the reference product if the FDA
approves a full BLA for the competing product containing the
sponsor’s own preclinical data and data from adequate and
well-controlled clinical trials to demonstrate the safety, purity
and potency of their product. We believe that any of our product
candidates approved as a biological product under a BLA should
qualify for the 12-year period of exclusivity. However, there is a
risk that this exclusivity could be shortened due to congressional
action or otherwise, or that the FDA will not consider our product
candidates to be reference products for competing products,
potentially creating the opportunity for competition sooner than
anticipated.
The commercial success of our product candidates will depend upon
the degree of market acceptance of such product candidates by
physicians, patients, healthcare payors and others in the medical
community.
Our product candidates may not be commercially successful. Even if
any of our product candidates receive regulatory approval, they may
not gain market acceptance among physicians, patients, healthcare
payors or the medical community. The commercial success of any of
our current or future product candidates will depend significantly
on the broad adoption and use of the resulting product by
physicians and patients for approved indications. The degree of
market acceptance of our products will depend on a number of
factors, including:
•
demonstration of clinical efficacy and safety compared to other
more-established products;
•
the indications for which our product candidates are
approved;
•
the limitation of our targeted patient population and other
limitations or warnings contained in any FDA-approved
labeling;
•
acceptance of a new drug for the relevant indication by healthcare
providers and their patients;
•
the pricing and cost-effectiveness of our products, as well as the
cost of treatment with our products in relation to alternative
treatments and therapies;
•
our ability to obtain and maintain sufficient third-party coverage
and adequate reimbursement from government healthcare programs,
including Medicare and Medicaid, private health insurers and other
third-party payors;
•
the willingness of patients to pay all, or a portion of,
out-of-pocket costs associated with our products in the absence of
sufficient third-party coverage and adequate
reimbursement;
•
any restrictions on the use of our products, and the prevalence and
severity of any adverse effects;
•
potential product liability claims;
•
the timing of market introduction of our products as well as
competitive drugs;
•
the effectiveness of our or any of our current or potential future
collaborators’ sales and marketing strategies; and
•
unfavorable publicity relating to the product, our company, or
product candidates or similar approved products or product
candidates in development by third parties.
94
If any product candidate is approved but does not achieve an
adequate level of acceptance by physicians, hospitals, healthcare
payors or patients, we may not generate sufficient revenue from
that product and may not become or remain profitable. Our efforts
to educate the medical community and third-party payors regarding
the benefits of our products may require significant resources and
may never be successful.
The successful commercialization of our product candidates, if
approved, will depend in part on the extent to which governmental
authorities and health insurers establish coverage, adequate
reimbursement levels and favorable pricing policies. Failure to
obtain or maintain coverage and adequate reimbursement for our
products could limit our ability to market those products and
decrease our ability to generate revenue.
The availability of coverage and the adequacy of reimbursement by
governmental healthcare programs such as Medicare and Medicaid,
private health insurers and other third-party payors are essential
for most patients to be able to afford prescription medications
such as our product candidates, if approved. Our ability to achieve
coverage and acceptable levels of reimbursement for our products by
third-party payors will have an effect on our ability to
successfully commercialize those products. Accordingly, we will
need to successfully implement a coverage and reimbursement
strategy for any approved product candidate. Even if we obtain
coverage for a given product by a third-party payor, the resulting
reimbursement payment rates may not be adequate or may require
co-payments that patients find unacceptably high. For products
administered under the supervision of a physician, obtaining
coverage and adequate reimbursement may be particularly difficult
because of the higher prices often associated with such drugs.
Additionally, separate reimbursement for the product itself or the
treatment or procedure in which the product is used may not be
available, which may impact physician utilization. We cannot be
sure that coverage and reimbursement in the United States, the
European Union or elsewhere will be available for any product that
we may develop, and any reimbursement that may become available may
be decreased or eliminated in the future.
Third-party payors increasingly are challenging prices charged for
biopharmaceutical products and services, and many third-party
payors may refuse to provide coverage and reimbursement for
particular drugs when an equivalent generic drug or a less
expensive therapy is available. It is possible that a third-party
payor may consider our products as substitutable and only offer to
reimburse patients for the less expensive product. Even if we are
successful in demonstrating improved efficacy or improved
convenience of administration with our products, pricing of
existing drugs may limit the amount we will be able to charge for
our products. These payors may deny or revoke the reimbursement
status of a given product or establish prices for new or existing
marketed products at levels that are too low to enable us to
realize an appropriate return on our investment in product
development. If reimbursement is not available or is available only
at limited levels, we may not be able to successfully commercialize
our products and may not be able to obtain a satisfactory financial
return on products that we may develop. In addition, in the event
that we develop companion diagnostic tests for use with our
products, once approved, such companion diagnostic tests will
require coverage and reimbursement separate and apart from the
coverage and reimbursement for their companion pharmaceutical or
biological products. Similar challenges to obtaining coverage and
reimbursement applicable to pharmaceutical or biological products
will apply to companion diagnostics tests.
There is significant uncertainty related to third-party payor
coverage and reimbursement of newly approved products. In the
United States, third-party payors, including private and
governmental payors, such as the Medicare and Medicaid programs,
play an important role in determining the extent to which new drugs
will be covered. Some third-party payors may require pre-approval
of coverage for new or innovative devices or drug therapies before
they will reimburse healthcare providers who use such therapies. It
is difficult to predict at this time what third-party payors will
decide with respect to the coverage and reimbursement for our
products.
Obtaining and maintaining reimbursement status is time-consuming,
costly and uncertain. The Medicare and Medicaid programs
increasingly are used as models for how private payors and other
governmental payors develop their coverage and reimbursement
policies for drugs. However, no uniform policy for coverage and
reimbursement for products exists among third-party payors in the
United States. Therefore, coverage and reimbursement for products
can differ significantly from payor to payor. As a result, the
coverage determination process is often a time-consuming and costly
process that will require us to provide scientific and clinical
support for the use of our products to each payor separately, with
no assurance that coverage and adequate reimbursement will be
applied consistently or obtained in the first instance.
Furthermore, rules and regulations regarding reimbursement change
frequently, in some cases at short notice, and we believe that
changes in these rules and regulations are likely.
Outside the United States, international operations are generally
subject to extensive governmental price controls and other market
regulations, and we believe the increasing emphasis on
cost-containment initiatives in Europe and other countries has and
will continue to put pressure on the pricing and usage of our
products. In many countries, the prices of
95
medical products are subject to varying price control mechanisms as
part of national health systems. Other countries allow companies to
fix their own prices for medical products but monitor and control
company profits. Additional foreign price controls or other changes
in pricing regulation could restrict the amount that we are able to
charge for our products. Accordingly, in markets outside the United
States, the reimbursement for our products may be reduced compared
with the United States and may be insufficient to generate
commercially reasonable revenue and profits.
Moreover, increasing efforts by governmental and third-party payors
in the United States and abroad to cap or reduce healthcare costs
may cause such organizations to limit both coverage and the level
of reimbursement for newly approved products and, as a result, they
may not cover or provide adequate payment for our products. We
expect to experience pricing pressures in connection with the sale
of any of our products due to the trend toward managed healthcare,
the increasing influence of health maintenance organizations and
additional legislative changes. The downward pressure on healthcare
costs in general, particularly prescription drugs and surgical
procedures and other treatments, has become very intense. As a
result, increasingly high barriers are being erected to the entry
of new products.
We face significant competition from entities that have developed
or may develop product candidates for cancer, including companies
developing novel treatments and technology platforms. If our
competitors develop technologies or product candidates more rapidly
than we do or their technologies are more effective, our business
and our ability to develop and successfully commercialize products
may be adversely affected.
Although the biotechnology and pharmaceutical industries, and the
oncology sector, are characterized by rapid evolution of
technologies, fierce competition, and strong defense of
intellectual property rights, we believe the most fearsome
competitor of all is cancer itself. As such, we view other
companies in this sector more as potential allies and collaborators
than as competitors, as we all have a common cause: to defeat
cancer. Many of the companies that are developing or marketing
treatments for cancer, including major pharmaceutical and
biotechnology companies that are working on therapies targeting the
RAS/MAPK pathway, are companies with whom we endeavor to
collaborate in our mission to erase cancer. That being said, our
commercial potential could be reduced or eliminated if other
companies develop and commercialize products that are safer, more
effective, have fewer or less severe side effects, are more
convenient or are less expensive than products that we may develop.
Our competitors have developed, are developing or may develop
products, product candidates and processes competitive with our
product candidates. Any product candidates that we successfully
develop and commercialize will compete with existing therapies and
new therapies that may become available in the future. We believe
that a significant number of products are currently under
development, and may become commercially available in the future,
for the treatment of indications for which we may attempt to
develop product candidates. In particular, there is intense
competition in the oncology field. Our competitors include larger
and better funded pharmaceutical, biopharmaceutical,
biotechnological and therapeutics companies. Moreover, we may also
compete with universities and other research institutions who may
be active in oncology research and could be in direct competition
with us. We also compete with these organizations to recruit
management, scientists and clinical development personnel, which
could negatively affect our level of expertise and our ability to
execute our business plan. We will also face competition in
establishing clinical trial sites, enrolling subjects for clinical
trials and in identifying and in-licensing new product candidates.
Smaller or early-stage companies may also prove to be significant
competitors, particularly through collaborative arrangements with
large and established companies.
If any of our product candidates are approved, they will compete
with small molecule therapies, biologics, cell-based therapies and
traditional chemotherapy, either approved or under development,
which are intended to treat the same indications that we are
targeting or may target, including through approaches that may
prove to be more effective, have fewer side effects, be less costly
to manufacture, be more convenient to administer or have other
advantages over our product candidates. In addition to competing
with other therapies targeting similar indications, there are
numerous other companies and academic institutions focused on
similar targets as our product candidates and/or different
scientific approaches to treating the same indications. We face
competition from such companies in seeking any future potential
collaborations to partner our product candidates, as well as
potentially competing commercially for any approved
products.
Specifically, there are also a number of pharmaceutical companies
with product candidates in development that target the nodes
involving the RAS/MAPK pathway. These include, among others, Amgen,
AstraZeneca, Black Diamond Therapeutics, BioMed Valley Discoveries,
Boehringer Ingelheim, Deciphera Pharmaceuticals, Eli Lilly, Jacobio
Pharmaceuticals (in collaboration with AbbVie), Janssen, Merck,
Mirati Therapeutics, Navire Pharma (a subsidiary of BridgeBio),
Novartis, Pfizer, Relay Therapeutics (in collaboration with
Genentech), Revolution Medicines, Roche/Genentech, Sanofi, and
Schrödinger (in collaboration with Bristol Myers
Squibb).
96
Many of our competitors have significantly greater financial,
technical, manufacturing, marketing, sales and supply resources or
experience than we do. If we successfully obtain approval for any
product candidate, we will face competition based on many different
factors, including the safety and effectiveness of our products,
the ease with which our products can be administered and the extent
to which patients accept relatively new routes of administration,
the timing and scope of regulatory approvals for these products,
the availability and cost of manufacturing, marketing and sales
capabilities, price, reimbursement coverage and patent position.
Competing products could present superior treatment alternatives,
including by being more effective, safer, more convenient, less
expensive or marketed and sold more effectively than any products
we may develop. Competitive products approaches may make any
products we develop obsolete or noncompetitive before we recover
the expense of developing and commercializing our product
candidates. If we are unable to compete effectively, our
opportunity to generate revenue from the sale of our products we
may develop, if approved, could be adversely affected.
The market opportunities for our product candidates may be limited
to patients who are ineligible for or have failed prior treatments
and may be small or different from our estimates.
Cancer therapies are defined by lines of therapy as well as by
treatment-naïve or previously-treated status. Often the initial
approval for a new therapy is in later lines and subsequent
approval in an earlier line may not be feasible. When cancer is
detected early enough, first line therapy is sometimes adequate to
cure the cancer or prolong life without a cure. Whenever first line
therapy, including targeted therapy, immunotherapy, chemotherapy,
hormone therapy, surgery or a combination of these, proves
unsuccessful, second line therapy may be administered. Second line
therapies often consist of additional chemotherapy, radiation,
antibody drugs, tumor targeted small molecules or a combination of
these. Third line therapies can include bone marrow
transplantation, antibody and small molecule targeted therapies,
more invasive forms of surgery and new technologies. In markets
with approved therapies, there is no guarantee that our product
candidates, even if approved, would be approved for second line or
first line therapy. This could limit our potential market
opportunity. In addition, we may have to conduct additional
clinical trials prior to gaining approval for second line or first
line therapy.
Our projections of both the number of people who have the cancers
we are targeting, as well as the subset of people with these
cancers in a position to receive later stage therapy and who have
the potential to benefit from treatment with our product
candidates, are based on our beliefs and estimates. These estimates
have been derived from a variety of sources, including the
scientific literature, publicly available clinical molecular
reports, patient foundations or market research, and may prove to
be incorrect. Further, new trials or information may change the
estimated incidence or prevalence of these cancers. The number of
patients in the United States and other major markets and elsewhere
may turn out to be lower than expected, patients may not be
otherwise amenable to treatment with our products or new patients
may become increasingly difficult to identify or gain access to,
all of which would adversely affect our results of operations and
our business. Further, even if we obtain significant market share
for our product candidates, because some of our potential target
populations are very small, we may never achieve profitability
despite obtaining such significant market share.
We currently have no marketing and sales organization and have no
experience as a company in commercializing products, and we may
have to invest significant resources to develop these capabilities.
If we are unable to establish marketing and sales capabilities or
enter into agreements with third parties to market and sell our
products, we may not be able to generate product
revenue.
We have no internal sales, marketing or distribution capabilities,
nor have we ever commercialized a product. If any of our product
candidates ultimately receives regulatory approval, we must build a
marketing and sales organization with technical expertise and
supporting distribution capabilities to commercialize each such
product in major markets, which will be expensive and
time-consuming, or collaborate with third parties that have direct
sales forces and established distribution systems, either to
augment our own sales force and distribution systems or in lieu of
our own sales force and distribution systems. We have no prior
experience as a company in the marketing, sale and distribution of
biopharmaceutical products and there are significant risks involved
in building and managing a sales organization, including our
ability to hire, retain and incentivize qualified individuals,
generate sufficient sales leads, provide adequate training to sales
and marketing personnel and effectively manage a geographically
dispersed sales and marketing team. Any failure or delay in the
development of our internal sales, marketing and distribution
capabilities would adversely impact the commercialization of these
products. We may not be able to enter into collaborations or hire
consultants or external service providers to assist us in sales,
marketing and distribution functions on acceptable financial terms,
or at all. In addition, our product revenue and our profitability,
if any, may be lower if we rely on third parties for these
functions than if we were to market, sell and distribute any
products that we develop ourselves. We likely will have little
control over such third parties, and any of them may fail to devote
the necessary resources and attention to sell and market
our
97
products effectively. If we are not successful in commercializing
our products, either on our own or through arrangements with one or
more third parties, we may not be able to generate any future
product revenue and we would incur significant additional
losses.
Our future growth may depend, in part, on our ability to operate in
foreign markets, where we would be subject to additional regulatory
burdens and other risks and uncertainties.
Our future growth may depend, in part, on our ability to develop
and commercialize our product candidates in foreign markets. We are
not permitted to market or promote any of our product candidates
before we receive regulatory approval from applicable regulatory
authorities in foreign markets, and we may never receive such
regulatory approvals for any of our product candidates. To obtain
separate regulatory approval in many other countries we must comply
with numerous and varying regulatory requirements regarding safety
and efficacy and governing, among other things, clinical trials,
commercial sales, pricing and distribution of our product
candidates. If we obtain regulatory approval of our product
candidates and ultimately commercialize our products in foreign
markets, we would be subject to additional risks and uncertainties,
including:
•
different regulatory requirements for approval of drugs in foreign
countries;
•
reduced protection for intellectual property rights;
•
the existence of additional third-party patent rights of potential
relevance to our business;
•
unexpected changes in tariffs, trade barriers and regulatory
requirements;
•
economic weakness, including inflation, or political instability in
particular foreign economies and markets;
•
compliance with tax, employment, immigration and labor laws for
employees living or traveling abroad;
•
foreign currency fluctuations, which could result in increased
operating expenses and reduced revenue, and other obligations
incident to doing business in another country;
•
foreign reimbursement, pricing and insurance regimes;
•
workforce uncertainty in countries where labor unrest is
common;
•
production shortages resulting from any events affecting raw
material supply or manufacturing capabilities abroad;
and
•
business interruptions resulting from geopolitical actions,
including war and terrorism, or natural disasters including
earthquakes, typhoons, floods and fires.
Risks related to our business operations and industry
Our operating results may fluctuate significantly, which makes our
future operating results difficult to predict and could cause our
operating results to fall below expectations or any guidance we may
provide.
Our quarterly and annual operating results may fluctuate
significantly, which makes it difficult for us to predict our
future operating results. These fluctuations may occur due to a
variety of factors, many of which are outside of our control,
including, but not limited to:
•
the timing and cost of, and level of investment in, research,
development, regulatory approval and commercialization activities
relating to our product candidates, which may change from time to
time;
•
the timing and success or failure of preclinical studies or
clinical trials for our product candidates or competing product
candidates, or any other change in the competitive landscape of our
industry, including consolidation among our competitors or
partners;
•
coverage and reimbursement policies with respect to our product
candidates, if approved, and potential future drugs that compete
with our products;
•
the cost of manufacturing our product candidates, which may vary
depending on the quantity of production and the terms of our
agreements with third-party manufacturers;
•
expenditures that we will or may incur to acquire, develop or
commercialize additional product candidates and technologies or
other assets;
98
•
the level of demand for any approved products, which may vary
significantly and be difficult to predict; and
•
future accounting pronouncements or changes in our accounting
policies;
The cumulative effects of these factors could result in large
fluctuations and unpredictability in our quarterly and annual
operating results. As a result, comparing our operating results on
a period-to-period basis may not be meaningful. Investors should
not rely on our past results as an indication of our future
performance.
This variability and unpredictability could also result in our
failing to meet the expectations of industry or financial analysts
or investors for any period. If our revenue or operating results
fall below the expectations of analysts or investors or below any
forecasts we may provide to the market, or if the forecasts we
provide to the market are below the expectations of analysts or
investors, the price of our common stock could decline
substantially. Such a stock price decline could occur even when we
have met any previously publicly stated revenue or earnings
guidance we may provide.
Our success is dependent on our ability to attract and retain
highly qualified management and other clinical and scientific
personnel.
Our success depends in part on our continued ability to attract,
retain, manage, and motivate highly qualified management, clinical,
and scientific personnel, and we face significant competition for
experienced personnel. We are highly dependent upon our senior
management, as well as our senior scientists and other members of
our management team. The loss of services of any of these
individuals could delay or prevent the successful development of
our product pipeline, initiation, or completion of our clinical
trials and preclinical studies or the commercialization of our
product candidates. Although we have executed employment agreements
or offer letters with each member of our senior management team,
these agreements are terminable at will with or without notice and,
therefore, we may not be able to retain their services as expected.
We do not currently maintain “key person” life insurance on the
lives of our executives or any of our employees. This lack of
insurance means that we may not have adequate compensation for the
loss of the services of these individuals.
We will need to expand and effectively manage our managerial,
operational, financial, and other resources in order to
successfully pursue our clinical development and commercialization
efforts. We may not be successful in maintaining our unique company
culture and continuing to attract or retain qualified management,
clinical, and scientific personnel in the future due to the intense
competition for qualified personnel among biopharmaceutical,
biotechnology, and other businesses, particularly in the San Diego
area. Our industry has experienced a high rate of turnover of
management personnel in recent years. If we are not able to
attract, integrate, retain, and motivate necessary personnel to
accomplish our business objectives, we may experience constraints
that will significantly impede the achievement of our development
objectives, our ability to raise additional capital, and our
ability to implement our business strategy.
We may encounter difficulties in managing our growth and expanding
our operations successfully.
We have substantially increased our organization from 30 employees
as of December 31, 2019 to 123 employees as of February 28, 2022.
As we continue development and pursue the potential
commercialization of our product candidates, as well as function as
a public company, we will need to expand our financial,
development, regulatory, manufacturing, marketing and sales
capabilities or contract with third parties to provide these
capabilities for us. As our operations expand, we expect that we
will need to manage additional relationships with various strategic
partners, suppliers and other third parties. Our future financial
performance and our ability to develop and commercialize our
product candidates and to compete effectively will depend, in part,
on our ability to manage any future growth effectively.
We are subject to various US federal, state and foreign healthcare
laws and regulations, which could increase compliance costs, and
our failure to comply with these laws and regulations could harm
our reputation, subject us to significant fines and liability or
otherwise adversely affect our business.
Our business operations and current and future arrangements with
investigators, healthcare professionals, consultants, third-party
payors, patient organizations and customers expose us to broadly
applicable foreign, federal and state fraud and abuse and other
healthcare laws and regulations. These laws may constrain the
business or financial arrangements and relationships through which
we conduct our operations, including how we research, market, sell
and distribute any products for which we obtain marketing approval.
Such laws include:
•
the federal Anti-Kickback Statute, which prohibits, among other
things, persons or entities from knowingly and willfully
soliciting, offering, receiving or providing any remuneration
(including any kickback, bribe or certain rebates), directly or
indirectly, overtly or covertly, in cash or in kind, in return for,
either the referral of an
99
individual or the purchase, lease, or order, or arranging for or
recommending the purchase, lease, or order of any good, facility,
item or service, for which payment may be made, in whole or in
part, under a federal healthcare program such as Medicare and
Medicaid. A person or entity does not need to have actual knowledge
of the federal Anti-Kickback Statute or specific intent to violate
it in order to have committed a violation;
•
the federal false claims laws, including the civil False Claims
Act, and civil monetary penalties laws, which prohibit, among other
things, individuals or entities from knowingly presenting, or
causing to be presented, to the federal government, claims for
payment or approval that are false or fraudulent, knowingly making,
using or causing to be made or used, a false record or statement
material to a false or fraudulent claim, or from knowingly making
or causing to be made a false statement to avoid, decrease or
conceal an obligation to pay money to the federal government. In
addition, the government may assert that a claim including items or
services resulting from a violation of the federal Anti-Kickback
Statute constitutes a false or fraudulent claim for purposes of the
civil False Claims Act;
•
the federal Health Insurance Portability and Accountability Act of
1996 (HIPAA), which imposes criminal and civil liability for, among
other things, knowingly and willfully executing, or attempting to
execute, a scheme to defraud any healthcare benefit program, or
knowingly and willfully falsifying, concealing or covering up a
material fact or making any materially false statement, in
connection with the delivery of, or payment for, healthcare
benefits, items or services. Similar to the federal Anti-Kickback
Statute, a person or entity does not need to have actual knowledge
of the statute or specific intent to violate it in order to have
committed a violation;
•
the federal Physician Payments Sunshine Act, which requires certain
manufacturers of drugs, devices, biologics and medical supplies for
which payment is available under Medicare, Medicaid or the
Children’s Health Insurance Program (with certain exceptions) to
report annually to the CMS, information related to payments and
other “transfers of value” made to physicians (defined to include
doctors, dentists, optometrists, podiatrists and chiropractors),
certain non-physician practitioners (physician assistants, nurse
practitioners, clinical nurse specialists, certified nurse
anesthetists, anesthesiology assistants and certified
nurse-midwives) and teaching hospitals, as well as ownership and
investment interests held by such healthcare professionals and
their immediate family members; and
•
analogous state and foreign laws and regulations, such as state
anti-kickback and false claims laws, may apply to sales or
marketing arrangements and claims involving healthcare items or
services reimbursed by non-governmental third-party payors,
including private insurers; some state laws require biotechnology
companies to comply with the biotechnology industry’s voluntary
compliance guidelines and the relevant compliance guidance
promulgated by the federal government and may require certain
biotechnology companies to report information related to payments
and other transfers of value to physicians and other healthcare
providers or marketing expenditures; some state laws that require
biotechnology companies to report information on the pricing of
certain drug products; and some state and local laws require the
registration or pharmaceutical sales representatives.
Efforts to ensure that our current and future business arrangements
with third parties will comply with applicable healthcare laws and
regulations will involve ongoing substantial costs. It is possible
that governmental authorities will conclude that our business
practices may not comply with current or future statutes,
regulations or case law involving applicable fraud and abuse or
other healthcare laws and regulations. If our operations are found
to be in violation of any of these laws or any other governmental
regulations that may apply to us, we may be subject to significant
penalties, including civil, criminal and administrative penalties,
damages, fines, disgorgement, imprisonment, exclusion from
participation in government funded healthcare programs, including
Medicare and Medicaid, integrity oversight and reporting
obligations, contractual damages, reputational harm, diminished
profits and future earnings and the curtailment or restructuring of
our operations. Defending against any such actions can be costly,
time-consuming and may require significant financial and personnel
resources. Therefore, even if we are successful in defending
against any such actions that may be brought against us, our
business may be impaired. Further, if any of the physicians or
other healthcare providers or entities with whom we expect to do
business is found to be not in compliance with applicable laws,
they may be subject to significant criminal, civil or
administrative sanctions, including exclusions from government
funded healthcare program.
100
Recently enacted legislation, future legislation and healthcare
reform measures may increase the difficulty and cost for us to
obtain marketing approval for and commercialize our product
candidates and may affect the prices we may set.
In the United States and some foreign jurisdictions, there have
been, and we expect there will continue to be, a number of
legislative and regulatory changes to the healthcare system,
including cost-containment measures that may reduce or limit
coverage and reimbursement for newly approved drugs and affect our
ability to profitably sell any product candidates for which we
obtain marketing approval. In particular, there have been and
continue to be a number of initiatives at the US federal and state
levels that seek to reduce healthcare costs and improve the quality
of healthcare.
For example, in March 2010, the ACA was enacted in the United
States. Among the provisions of the ACA of importance to our
potential product candidates, the ACA: established an annual,
nondeductible fee on any entity that manufactures or imports
specified branded prescription drugs and biologic agents; extended
manufacturers’ Medicaid rebate liability to covered drugs dispensed
to individuals who are enrolled in Medicaid managed care
organizations; expands eligibility criteria for Medicaid programs;
expanded the entities eligible for discounts under the Public
Health Service pharmaceutical pricing program; increased the
statutory minimum rebates a manufacturer must pay under the
Medicaid Drug Rebate Program; creates a new Medicare Part D
coverage gap discount program; established a new Patient-Centered
Outcomes Research Institute to oversee, identify priorities in and
conduct comparative clinical effectiveness research, along with
funding for such research; and established a Center for Medicare
and Medicaid Innovation at CMS to test innovative payment and
service delivery models to lower Medicare and Medicaid
spending.
There have been executive, judicial and Congressional challenges to
certain aspects of the ACA. On June 17, 2021, the US Supreme Court
dismissed the most recent judicial challenge to the ACA brought by
several states without specifically ruling on the constitutionality
of the ACA. Prior to the Supreme Court’s decision, President Biden
issued an executive order to initiate a special enrollment period
from February 15, 2021 through August 15, 2021 for purposes of
obtaining health insurance coverage through the ACA marketplace.
The executive order also instructed certain governmental agencies
to review and reconsider their existing policies and rules that
limit access to healthcare, including among others, reexamining
Medicaid demonstration projects and waiver programs that include
work requirements, and policies that create unnecessary barriers to
obtaining access to health insurance coverage through Medicaid or
the ACA. It is unclear how the healthcare reform measures of the
Biden administration will impact our business.
In addition, other legislative changes have been proposed and
adopted since the ACA was enacted. On August 2, 2011, the Budget
Control Act of 2011 was signed into law, which, among other things,
resulted in reductions to Medicare payments to providers of 2% per
fiscal year, which went into effect on April 1, 2013 and, due to
subsequent legislative amendments to the statute, will remain in
effect through 2030, with the exception of a temporary suspension
from May 1, 2020 through March 31, 2022, unless additional
Congressional action is taken. On January 2, 2013, the American
Taxpayer Relief Act of 2012 was signed into law, which, among other
things, reduced Medicare payments to several providers, including
hospitals, and increased the statute of limitations period for the
government to recover overpayments to providers from three to five
years.
Further, there has been heightened governmental scrutiny in the
United States of pharmaceutical pricing practices in light of the
rising cost of prescription drugs. Such scrutiny has resulted in
several recent congressional inquiries and proposed and enacted
federal and state legislation designed to, among other things,
bring more transparency to product pricing, review the relationship
between pricing and manufacturer patient programs, and reform
government program reimbursement methodologies for products. At the
federal level, the Trump administration used several means to
propose or implement drug pricing reform, including through federal
budget proposals, executive orders and policy initiatives. It is
unclear whether the Biden administration will work to reverse these
measures or pursue similar policy initiatives. Further, it is
possible that additional governmental action is taken in response
to the COVID-19 pandemic.
At the state level, legislatures have increasingly passed
legislation and implemented regulations designed to control
pharmaceutical and biological product pricing, including price or
patient reimbursement constraints, discounts, restrictions on
certain product access and marketing cost disclosure and
transparency measures, and, in some cases, designed to encourage
importation from other countries and bulk purchasing. Legally
mandated price controls on payment amounts by third-party payors or
other restrictions could harm our business, results of operations,
financial condition and prospects. In addition, regional healthcare
authorities and individual hospitals are increasingly using bidding
procedures to determine what pharmaceutical products and which
suppliers will be included in their prescription drug and other
healthcare programs. This could reduce the ultimate demand for our
product candidates, if approved, or put pressure on our product
pricing, which could negatively affect our business, results of
operations, financial condition and prospects.
101
We expect that the ACA, these new laws and other healthcare reform
measures that may be adopted in the future may result in additional
reductions in Medicare and other healthcare funding, more rigorous
coverage criteria, new payment methodologies and additional
downward pressure on the price that we receive for any approved
product. Any reduction in reimbursement from Medicare or other
government programs may result in a similar reduction in pa