bbio-10k_20201231.htm
false FY 0001743881 --12-31 0 true true true true true true true true true true true P5D P5D 0.8333 true true us-gaap:AssetsNoncurrent P3Y 0 2027 P7Y 0.8333 P3Y P6M P4Y us-gaap:LicenseMember us-gaap:LicenseMember us-gaap:LicenseMember 12.0 12.0 0.1 1.6 P1Y3M P8M1D 80.0 60.0 1.16 2.34 84.0 84.0 14.3 13.1 P6Y3M P5Y3M 95.0 68.0 1.53 2.46 P5Y1M6D P5Y1M6D bbio:OtherAccruedLiabilitiesCurrentMember us-gaap:OtherNoncurrentLiabilitiesMember P9Y7M6D P8Y9M18D P8Y2M12D P8Y4M24D P8Y1M6D P4M24D P5Y P4M24D P5Y P6Y29D P7M24D P6Y29D 0.363 0.325 0.364 0.464 0.476 0.375 0.0031 0.0013 0.0169 0.0150 0.0157 0.0186 P1Y6M 0.480 0.400 P9M P1Y6M 0.490 0.490 0.0234 0.0170 0.0256 0.0256 P6M P6M P5M23D P8Y9M7D P8Y5M15D P7Y10M9D P8Y5M15D P6Y21D P6Y25D P6Y29D 0001743881 2020-01-01 2020-12-31 xbrli:shares 0001743881 2021-02-19 iso4217:USD 0001743881 2020-06-30 0001743881 2020-12-31 0001743881 2019-12-31 iso4217:USD xbrli:shares 0001743881 2019-01-01 2019-12-31 0001743881 2018-01-01 2018-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2017-12-31 0001743881 us-gaap:CommonStockMember 2017-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2017-12-31 0001743881 us-gaap:RetainedEarningsMember 2017-12-31 0001743881 us-gaap:ParentMember 2017-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2017-12-31 0001743881 2017-12-31 0001743881 us-gaap:CommonStockMember us-gaap:StockCompensationPlanMember 2018-01-01 2018-12-31 0001743881 us-gaap:ParentMember us-gaap:StockCompensationPlanMember 2018-01-01 2018-12-31 0001743881 us-gaap:StockCompensationPlanMember 2018-01-01 2018-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2018-01-01 2018-12-31 0001743881 us-gaap:ParentMember 2018-01-01 2018-12-31 0001743881 us-gaap:CommonStockMember bbio:FourPointTwoNinePerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:TreasuryStockMember bbio:FourPointTwoNinePerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember bbio:FourPointTwoNinePerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:ParentMember bbio:FourPointTwoNinePerShareMember 2018-01-01 2018-12-31 0001743881 bbio:FourPointTwoNinePerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:CommonStockMember bbio:NinePointEightTwoPerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:TreasuryStockMember bbio:NinePointEightTwoPerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember bbio:NinePointEightTwoPerShareMember 2018-01-01 2018-12-31 0001743881 us-gaap:ParentMember bbio:NinePointEightTwoPerShareMember 2018-01-01 2018-12-31 0001743881 bbio:NinePointEightTwoPerShareMember 2018-01-01 2018-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2018-01-01 2018-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2018-01-01 2018-12-31 0001743881 us-gaap:RetainedEarningsMember 2018-01-01 2018-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2018-12-31 0001743881 us-gaap:CommonStockMember 2018-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2018-12-31 0001743881 us-gaap:RetainedEarningsMember 2018-12-31 0001743881 us-gaap:ParentMember 2018-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2018-12-31 0001743881 2018-12-31 0001743881 us-gaap:CommonStockMember us-gaap:StockCompensationPlanMember 2019-01-01 2019-12-31 0001743881 us-gaap:ParentMember us-gaap:StockCompensationPlanMember 2019-01-01 2019-12-31 0001743881 us-gaap:StockCompensationPlanMember 2019-01-01 2019-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2019-01-01 2019-12-31 0001743881 us-gaap:ParentMember 2019-01-01 2019-12-31 0001743881 us-gaap:CommonStockMember us-gaap:IPOMember 2019-01-01 2019-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember us-gaap:IPOMember 2019-01-01 2019-12-31 0001743881 us-gaap:ParentMember us-gaap:IPOMember 2019-01-01 2019-12-31 0001743881 us-gaap:IPOMember 2019-01-01 2019-12-31 0001743881 us-gaap:CommonStockMember us-gaap:EmployeeStockMember 2019-01-01 2019-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember us-gaap:EmployeeStockMember 2019-01-01 2019-12-31 0001743881 us-gaap:ParentMember us-gaap:EmployeeStockMember 2019-01-01 2019-12-31 0001743881 us-gaap:EmployeeStockMember 2019-01-01 2019-12-31 0001743881 us-gaap:AccumulatedOtherComprehensiveIncomeMember 2019-01-01 2019-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2019-01-01 2019-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2019-01-01 2019-12-31 0001743881 us-gaap:RetainedEarningsMember 2019-01-01 2019-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2019-12-31 0001743881 us-gaap:CommonStockMember 2019-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2019-12-31 0001743881 us-gaap:AccumulatedOtherComprehensiveIncomeMember 2019-12-31 0001743881 us-gaap:RetainedEarningsMember 2019-12-31 0001743881 us-gaap:ParentMember 2019-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2019-12-31 0001743881 us-gaap:CommonStockMember us-gaap:StockCompensationPlanMember 2020-01-01 2020-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember us-gaap:StockCompensationPlanMember 2020-01-01 2020-12-31 0001743881 us-gaap:ParentMember us-gaap:StockCompensationPlanMember 2020-01-01 2020-12-31 0001743881 us-gaap:StockCompensationPlanMember 2020-01-01 2020-12-31 0001743881 us-gaap:CommonStockMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 us-gaap:ParentMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 us-gaap:NoncontrollingInterestMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2020-01-01 2020-12-31 0001743881 us-gaap:ParentMember 2020-01-01 2020-12-31 0001743881 us-gaap:CommonStockMember 2020-01-01 2020-12-31 0001743881 us-gaap:TreasuryStockMember 2020-01-01 2020-12-31 0001743881 us-gaap:CommonStockMember bbio:SatisfyTaxWithholdingMember 2020-01-01 2020-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember bbio:SatisfyTaxWithholdingMember 2020-01-01 2020-12-31 0001743881 us-gaap:ParentMember bbio:SatisfyTaxWithholdingMember 2020-01-01 2020-12-31 0001743881 bbio:SatisfyTaxWithholdingMember 2020-01-01 2020-12-31 0001743881 us-gaap:AccumulatedOtherComprehensiveIncomeMember 2020-01-01 2020-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2020-01-01 2020-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2020-01-01 2020-12-31 0001743881 us-gaap:RetainedEarningsMember 2020-01-01 2020-12-31 0001743881 bbio:RedeemableConvertibleNoncontrollingInterestsMember 2020-12-31 0001743881 us-gaap:CommonStockMember 2020-12-31 0001743881 us-gaap:TreasuryStockMember 2020-12-31 0001743881 us-gaap:AdditionalPaidInCapitalMember 2020-12-31 0001743881 us-gaap:AccumulatedOtherComprehensiveIncomeMember 2020-12-31 0001743881 us-gaap:RetainedEarningsMember 2020-12-31 0001743881 us-gaap:ParentMember 2020-12-31 0001743881 us-gaap:NoncontrollingInterestMember 2020-12-31 0001743881 us-gaap:CommonStockMember bbio:FourPointTwoNinePerShareMember 2018-12-31 0001743881 us-gaap:CommonStockMember bbio:NinePointEightTwoPerShareMember 2018-12-31 0001743881 us-gaap:CommonStockMember us-gaap:IPOMember 2019-12-31 0001743881 bbio:LEOCallOptionMember 2020-01-01 2020-12-31 0001743881 bbio:TwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-01-01 2020-12-31 0001743881 bbio:HerculesCapitalIncMember 2020-01-01 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:DebtInstrumentTrancheAMember 2020-01-01 2020-12-31 0001743881 bbio:MarketBasedRestrictedStockUnitsMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenEmployeeStockPurchasePlanMember 2020-01-01 2020-12-31 0001743881 srt:SubsidiariesMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandEighteenEmployeeStockPurchasePlanMember 2020-01-01 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember 2020-01-01 2020-12-31 0001743881 bbio:EmployeeMember bbio:EidosTherapeuticsIncMember 2020-01-01 2020-12-31 xbrli:pure 0001743881 srt:MinimumMember 2020-01-01 2020-12-31 0001743881 bbio:BiotechCompanyMember us-gaap:CommonStockMember 2020-12-31 0001743881 bbio:BiotechCompanyMember us-gaap:CommonStockMember 2019-12-31 0001743881 bbio:TwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-12-31 0001743881 us-gaap:PrepaidExpensesAndOtherCurrentAssetsMember 2020-12-31 0001743881 us-gaap:OtherAssetsMember 2020-12-31 0001743881 us-gaap:OtherAssetsMember 2019-12-31 0001743881 us-gaap:OtherAssetsMember 2018-12-31 0001743881 us-gaap:FurnitureAndFixturesMember srt:MinimumMember 2020-01-01 2020-12-31 0001743881 us-gaap:FurnitureAndFixturesMember srt:MaximumMember 2020-01-01 2020-12-31 0001743881 bbio:LaboratoryAndMachineryEquipmentMember srt:MinimumMember 2020-01-01 2020-12-31 0001743881 bbio:LaboratoryAndMachineryEquipmentMember srt:MaximumMember 2020-01-01 2020-12-31 0001743881 us-gaap:LeaseholdImprovementsMember 2020-01-01 2020-12-31 bbio:Segment 0001743881 us-gaap:GeographicConcentrationRiskMember us-gaap:SalesRevenueNetMember country:CN 2020-01-01 2020-12-31 0001743881 us-gaap:GeographicConcentrationRiskMember us-gaap:SalesRevenueNetMember country:US bbio:AlexionAgreementsMember 2019-01-01 2019-12-31 0001743881 us-gaap:GeographicConcentrationRiskMember us-gaap:SalesRevenueNetMember country:CN bbio:BiotechCompanyMember 2019-01-01 2019-12-31 0001743881 us-gaap:GeographicConcentrationRiskMember us-gaap:SalesRevenueNetMember 2018-01-01 2018-12-31 0001743881 srt:MaximumMember srt:SubsidiariesMember bbio:ManagementIncentiveUnitsAndCommonUnitsMember 2020-01-01 2020-12-31 0001743881 us-gaap:AccountingStandardsUpdate201602Member 2020-01-01 0001743881 us-gaap:AccountingStandardsUpdate201602Member bbio:BuildToSuitLeaseAssetMember 2020-01-01 2020-01-01 0001743881 us-gaap:AccountingStandardsUpdate201602Member bbio:OtherAccruedLiabilitiesMember 2020-01-01 2020-01-01 0001743881 us-gaap:AccountingStandardsUpdate201602Member us-gaap:ConstructionInProgressMember 2020-01-01 2020-01-01 0001743881 us-gaap:AccountingStandardsUpdate201602Member bbio:BuildToSuitLeaseLiabilityMember 2020-01-01 2020-01-01 0001743881 us-gaap:AccountingStandardsUpdate201602Member 2020-12-31 0001743881 us-gaap:AccountingStandardsUpdate201613Member 2020-12-31 0001743881 us-gaap:AccountingStandardsUpdate201813Member 2020-12-31 0001743881 us-gaap:AccountingStandardsUpdate201815Member 2020-12-31 0001743881 us-gaap:AccountingStandardsUpdate201912Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:MoneyMarketFundsMember 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:MoneyMarketFundsMember us-gaap:FairValueInputsLevel1Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:USTreasuryBillSecuritiesMember 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:USTreasuryBillSecuritiesMember us-gaap:FairValueInputsLevel2Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:USTreasurySecuritiesMember 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:USTreasurySecuritiesMember us-gaap:FairValueInputsLevel2Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CommercialPaperMember 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CommercialPaperMember us-gaap:FairValueInputsLevel2Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CorporateDebtSecuritiesMember 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CorporateDebtSecuritiesMember us-gaap:FairValueInputsLevel2Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:FairValueInputsLevel2Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:FairValueInputsLevel3Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:FairValueInputsLevel1Member 2020-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:MoneyMarketFundsMember 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:MoneyMarketFundsMember us-gaap:FairValueInputsLevel1Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:RepurchaseAgreementsMember 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:RepurchaseAgreementsMember us-gaap:FairValueInputsLevel1Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:FairValueInputsLevel1Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:USTreasurySecuritiesMember 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:USTreasurySecuritiesMember us-gaap:FairValueInputsLevel2Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CommercialPaperMember 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CommercialPaperMember us-gaap:FairValueInputsLevel2Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CorporateDebtSecuritiesMember 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:CorporateDebtSecuritiesMember us-gaap:FairValueInputsLevel2Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:FairValueInputsLevel2Member 2019-12-31 0001743881 us-gaap:FairValueMeasurementsRecurringMember us-gaap:FairValueInputsLevel3Member 2019-12-31 0001743881 bbio:MeasurementInputProbabilityOfMilestoneAchievementMember srt:MinimumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 bbio:MeasurementInputProbabilityOfMilestoneAchievementMember srt:MaximumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 bbio:MeasurementInputProbabilityOfMilestoneAchievementMember srt:MinimumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 bbio:MeasurementInputProbabilityOfMilestoneAchievementMember srt:MaximumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 us-gaap:MeasurementInputDiscountRateMember srt:MinimumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:MeasurementInputDiscountRateMember srt:MaximumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:MeasurementInputDiscountRateMember srt:MinimumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 us-gaap:MeasurementInputDiscountRateMember srt:MaximumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 srt:MinimumMember bbio:LEOCallOptionMember 2020-01-01 2020-12-31 0001743881 srt:MaximumMember bbio:LEOCallOptionMember 2020-01-01 2020-12-31 0001743881 srt:MinimumMember bbio:LEOCallOptionMember 2019-01-01 2019-12-31 0001743881 srt:MaximumMember bbio:LEOCallOptionMember 2019-01-01 2019-12-31 0001743881 us-gaap:MeasurementInputPriceVolatilityMember srt:MinimumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:MeasurementInputPriceVolatilityMember srt:MaximumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:MeasurementInputPriceVolatilityMember srt:MinimumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 us-gaap:MeasurementInputPriceVolatilityMember srt:MaximumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 us-gaap:MeasurementInputRiskFreeInterestRateMember srt:MinimumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:MeasurementInputRiskFreeInterestRateMember srt:MaximumMember bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:MeasurementInputRiskFreeInterestRateMember srt:MinimumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 us-gaap:MeasurementInputRiskFreeInterestRateMember srt:MaximumMember bbio:LEOCallOptionMember 2019-12-31 0001743881 bbio:LEOCallOptionMember 2018-01-01 2018-12-31 0001743881 bbio:LEOCallOptionMember 2018-12-31 0001743881 bbio:LEOCallOptionMember 2019-01-01 2019-12-31 0001743881 bbio:LEOCallOptionMember 2019-12-31 0001743881 bbio:LEOCallOptionMember 2020-12-31 0001743881 us-gaap:USTreasurySecuritiesMember srt:MaximumMember 2020-12-31 0001743881 us-gaap:MoneyMarketFundsMember 2020-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:USTreasuryBillSecuritiesMember 2020-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:USTreasurySecuritiesMember 2020-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:CommercialPaperMember 2020-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:CorporateDebtSecuritiesMember 2020-12-31 0001743881 us-gaap:ShortTermInvestmentsMember 2020-12-31 0001743881 us-gaap:MoneyMarketFundsMember 2019-12-31 0001743881 us-gaap:RepurchaseAgreementsMember 2019-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:USTreasurySecuritiesMember 2019-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:CommercialPaperMember 2019-12-31 0001743881 us-gaap:ShortTermInvestmentsMember us-gaap:CorporateDebtSecuritiesMember 2019-12-31 0001743881 us-gaap:ShortTermInvestmentsMember 2019-12-31 0001743881 bbio:LongTermMarketableSecuritiesMember us-gaap:USTreasurySecuritiesMember 2019-12-31 0001743881 bbio:LongTermMarketableSecuritiesMember us-gaap:CorporateDebtSecuritiesMember 2019-12-31 0001743881 bbio:LongTermMarketableSecuritiesMember 2019-12-31 0001743881 us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2016-04-01 2016-04-30 0001743881 bbio:EidosTherapeuticsIncMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember 2018-02-28 0001743881 bbio:EidosTherapeuticsIncMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember 2018-02-01 2018-02-28 0001743881 bbio:EidosTherapeuticsIncMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember 2018-03-31 0001743881 bbio:EidosTherapeuticsIncMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember 2018-03-01 2018-03-31 0001743881 us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2018-05-01 2018-05-31 0001743881 us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2018-06-01 2018-06-30 0001743881 us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2018-06-22 2020-12-31 0001743881 us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2019-05-01 2019-05-31 0001743881 us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2019-07-01 2019-07-31 0001743881 us-gaap:CommonStockMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2019-09-01 2019-09-30 0001743881 bbio:AtTheMarketSalesAgreementMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2019-08-02 0001743881 bbio:AtTheMarketSalesAgreementMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2019-08-02 0001743881 bbio:AtTheMarketSalesAgreementMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2019-01-01 2019-12-31 0001743881 bbio:AtTheMarketSalesAgreementMember us-gaap:VariableInterestEntityPrimaryBeneficiaryMember bbio:EidosTherapeuticsIncMember 2020-02-01 2020-02-29 0001743881 bbio:EidosTherapeuticsIncMember bbio:AgreementAndPlanOfMergerMember 2020-10-05 0001743881 bbio:EidosTherapeuticsIncMember srt:MinimumMember bbio:AgreementAndPlanOfMergerMember 2020-10-05 0001743881 bbio:EidosTherapeuticsIncMember srt:MaximumMember bbio:AgreementAndPlanOfMergerMember 2020-10-05 0001743881 bbio:EidosTherapeuticsIncMember bbio:AgreementAndPlanOfMergerMember 2020-10-05 2020-10-05 0001743881 bbio:AgreementAndPlanOfMergerMember 2020-10-05 2020-10-05 0001743881 bbio:BridgeBioPharmaLimitedLiabilityCompanyMember bbio:EntitiesAffiliatedWithPerceptiveLifeSciencesMasterFundLtdMember 2019-10-31 0001743881 bbio:BridgeBioPharmaLimitedLiabilityCompanyMember bbio:EntitiesAffiliatedWithPerceptiveLifeSciencesMasterFundLtdMember 2019-12-31 0001743881 bbio:BridgeBioPharmaLimitedLiabilityCompanyMember bbio:EntitiesAffiliatedWithPerceptiveLifeSciencesMasterFundLtdMember 2019-10-01 2019-10-31 0001743881 bbio:PellePharmIncMember 2015-07-31 0001743881 srt:MinimumMember bbio:PellePharmIncMember 2015-07-01 2016-12-31 0001743881 bbio:PellePharmIncMember 2018-11-19 2018-11-19 0001743881 bbio:PellePharmIncMember 2018-11-19 0001743881 bbio:PellePharmIncMember us-gaap:CommonStockMember 2018-11-19 0001743881 bbio:PellePharmIncMember us-gaap:PreferredStockMember 2018-11-19 0001743881 us-gaap:EquityMethodInvestmentsMember us-gaap:CommonStockMember bbio:PellePharmIncMember 2018-11-19 0001743881 bbio:PellePharmIncMember 2019-01-01 2019-12-31 0001743881 bbio:PellePharmIncMember 2018-11-20 2018-12-31 0001743881 bbio:PellePharmIncMember 2019-12-31 0001743881 bbio:PellePharmIncMember 2018-12-31 0001743881 bbio:PellePharmIncMember 2019-03-31 0001743881 bbio:PellePharmIncMember 2019-04-01 2019-12-31 0001743881 bbio:PellePharmIncMember 2020-01-01 2020-12-31 0001743881 bbio:PellePharmIncMember 2018-01-01 2018-12-31 0001743881 bbio:OriginBiosciencesIncMember 2018-06-01 2018-06-30 0001743881 us-gaap:InProcessResearchAndDevelopmentMember bbio:OriginBiosciencesIncMember 2018-01-01 2018-12-31 0001743881 srt:MaximumMember bbio:OriginBiosciencesIncMember 2018-01-01 2018-12-31 0001743881 srt:MaximumMember bbio:OriginBiosciencesIncMember 2018-12-31 0001743881 bbio:QEDTherapeuticsIncMember 2018-01-01 2018-01-31 0001743881 us-gaap:SeriesAPreferredStockMember bbio:QEDTherapeuticsIncMember 2018-01-01 2018-01-31 0001743881 us-gaap:SeriesAPreferredStockMember bbio:QEDTherapeuticsIncMember 2018-01-31 0001743881 us-gaap:InProcessResearchAndDevelopmentMember bbio:QEDTherapeuticsIncMember 2018-01-01 2018-12-31 0001743881 srt:MaximumMember bbio:QEDTherapeuticsIncMember 2018-12-31 0001743881 srt:MaximumMember 2020-12-31 0001743881 bbio:TwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-03-09 0001743881 bbio:TwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-03-09 2020-03-09 bbio:TradingDay 0001743881 bbio:TwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember srt:MaximumMember 2020-03-09 2020-03-09 0001743881 bbio:PrincipalValueAndInterestOnTwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-12-31 0001743881 bbio:InterestAdjustOnTwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-12-31 0001743881 bbio:PrincipalValueOfTwoPointFiveZeroPercentageConvertibleSeniorNotesDueTwoThousandAndTwentySevenMember 2020-12-31 0001743881 bbio:CappedCallTransactionsMember 2020-03-03 2020-03-04 0001743881 bbio:CappedCallTransactionsMember 2020-03-04 0001743881 bbio:ShareRepurchaseTransactionsMember 2020-03-03 2020-03-04 0001743881 bbio:ShareRepurchaseTransactionsMember 2020-03-04 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2018-06-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2018-06-01 2018-06-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember us-gaap:PrimeRateMember 2018-06-01 2018-06-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember us-gaap:PrimeRateMember 2018-01-01 2018-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2018-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2018-12-01 2018-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember us-gaap:PrimeRateMember 2018-12-01 2018-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember us-gaap:PrimeRateMember 2018-12-31 0001743881 bbio:HerculesCapitalIncMember 2018-06-30 0001743881 bbio:HerculesCapitalIncMember 2018-12-31 0001743881 bbio:DebtInstrumentTrancheThreeMember bbio:HerculesCapitalIncMember 2019-05-31 0001743881 bbio:HerculesCapitalIncMember 2019-07-01 2019-07-31 0001743881 srt:MinimumMember us-gaap:PaymentInKindPIKNoteMember bbio:HerculesCapitalIncMember 2019-07-01 2019-07-31 0001743881 bbio:HerculesCapitalIncMember 2019-07-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember us-gaap:PrimeRateMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember us-gaap:PrimeRateMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember us-gaap:PrimeRateMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember us-gaap:PrimeRateMember 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember us-gaap:PrimeRateMember 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember us-gaap:PrimeRateMember 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember us-gaap:PrimeRateMember bbio:DebtInstrumentTrancheOneMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2020-04-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2020-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheOneMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember us-gaap:PrimeRateMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2020-04-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2020-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheTwoMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember us-gaap:PrimeRateMember bbio:DebtInstrumentTrancheThreeMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember 2020-04-30 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember 2020-12-31 0001743881 bbio:HerculesCapitalIncMember bbio:DebtInstrumentTrancheThreeMember 2020-04-01 2020-04-30 0001743881 bbio:HerculesCapitalIncMember 2020-04-30 0001743881 bbio:HerculesCapitalIncMember 2020-12-15 0001743881 bbio:HerculesCapitalIncMember 2020-12-31 0001743881 bbio:HerculesCapitalIncMember us-gaap:CollateralPledgedMember 2020-12-31 0001743881 bbio:HerculesCapitalIncMember 2019-01-01 2019-12-31 0001743881 bbio:HerculesCapitalIncMember 2018-01-01 2018-12-31 0001743881 bbio:HerculesTermLoanMember 2020-12-31 0001743881 bbio:HerculesTermLoanMember 2019-12-31 0001743881 bbio:PrincipalValueInterestOnTermLoansAndFinalEndOfTermPaymentsOfHerculesTermLoanMember 2020-12-31 0001743881 bbio:InterestAdjustOnHerculesTermLoanMember 2020-12-31 0001743881 bbio:FinalEndOfTermPaymentsOfHerculesTermLoanAdjustMember 2020-12-31 0001743881 bbio:PrincipalValueOfHerculesTermLoanMember 2020-12-31 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheBLoanMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheCLoanMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember 2019-11-12 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheBLoanMember bbio:EidosTherapeuticsIncMember 2019-11-12 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheCLoanMember bbio:EidosTherapeuticsIncMember 2019-11-12 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheBLoanMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2020-10-31 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember us-gaap:PrimeRateMember 2019-11-12 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember us-gaap:PrimeRateMember 2020-01-01 2020-12-31 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember srt:MinimumMember 2019-11-12 2019-11-13 0001743881 bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:EidosTherapeuticsIncMember srt:MaximumMember 2019-11-12 2019-11-13 0001743881 bbio:EidosTherapeuticsIncMember bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:ScenarioOneMember 2019-11-12 2019-11-13 0001743881 bbio:EidosTherapeuticsIncMember bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember bbio:ScenarioTwoMember 2019-11-12 2019-11-13 0001743881 bbio:EidosTherapeuticsIncMember bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember 2020-01-01 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember 2019-01-01 2019-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:SiliconValleyBankAndHerculesLoanMember bbio:TrancheALoanMember 2019-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:PrincipalValueAndInterestOnTermLoanOfSiliconValleyBankAndHerculesTermLoanMember bbio:DebtInstrumentTrancheAMember 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:InterestAdjustOnSiliconValleyBankAndHerculesTermLoanMember bbio:DebtInstrumentTrancheAMember 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:PrincipalValueOfSiliconValleyBankAndHerculesTermLoanMember bbio:DebtInstrumentTrancheAMember 2020-12-31 0001743881 bbio:QEDTherapeuticsIncMember bbio:EntitiesAffiliatedWithPerceptiveLifeSciencesMasterFundLtdMember 2019-10-01 2019-10-31 0001743881 bbio:QEDTherapeuticsIncMember bbio:EntitiesAffiliatedWithPerceptiveLifeSciencesMasterFundLtdMember srt:MaximumMember 2019-10-01 2019-10-31 0001743881 us-gaap:LicenseMember 2019-01-01 2019-12-31 0001743881 bbio:LianBioMember us-gaap:WarrantMember us-gaap:OtherNonoperatingIncomeExpenseMember 2020-01-01 2020-12-31 0001743881 bbio:LianBioMember 2020-01-01 2020-12-31 0001743881 bbio:LianBioMember bbio:LicenseAgreementMember 2020-08-01 2020-08-31 0001743881 bbio:LianBioMember srt:MaximumMember bbio:LicenseAgreementMember 2020-08-01 2020-08-31 0001743881 us-gaap:LicenseMember 2020-01-01 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionLicenseAgreementsMember 2019-09-01 2019-09-30 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionAgreementsMember us-gaap:CommonStockMember 2019-09-01 2019-09-30 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionAgreementsMember us-gaap:CommonStockMember 2019-09-30 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionAgreementsMember us-gaap:CommonStockMember exch:XNGS 2019-09-30 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionLicenseAgreementsMember 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionAgreementsMember 2019-09-01 2019-09-30 0001743881 bbio:EidosTherapeuticsIncMember bbio:AlexionLicenseAgreementsMember srt:MaximumMember 2020-12-31 0001743881 us-gaap:LicenseMember bbio:EidosTherapeuticsIncMember bbio:AlexionAgreementsMember 2020-01-01 2020-12-31 0001743881 us-gaap:LicenseMember bbio:ClinicalSupplyAgreementMember 2020-01-01 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:StanfordLicenseAgreementMember bbio:LelandStanfordJuniorUniversityMember 2017-03-01 2017-03-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:StanfordLicenseAgreementMember bbio:LelandStanfordJuniorUniversityMember 2016-08-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:StanfordLicenseAgreementMember bbio:LelandStanfordJuniorUniversityMember 2020-01-01 2020-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:StanfordLicenseAgreementMember bbio:LelandStanfordJuniorUniversityMember 2019-01-01 2019-12-31 0001743881 bbio:EidosTherapeuticsIncMember bbio:StanfordLicenseAgreementMember bbio:LelandStanfordJuniorUniversityMember 2018-01-01 2018-12-31 0001743881 us-gaap:LicenseAndServiceMember bbio:EidosTherapeuticsIncMember bbio:AlexionAgreementsMember bbio:LelandStanfordJuniorUniversityMember 2019-01-01 2019-12-31 0001743881 bbio:RegentsOfUniversityOfCaliforniaMember bbio:TheRegentsOfTheUniversityOfCaliforniaLicenseAgreementMember bbio:TheRasIncMember 2016-09-01 2016-09-30 0001743881 bbio:RegentsOfUniversityOfCaliforniaMember bbio:TheRegentsOfTheUniversityOfCaliforniaLicenseAgreementMember bbio:TheRasIncMember 2016-09-30 0001743881 bbio:RegentsOfUniversityOfCaliforniaMember bbio:TheRegentsOfTheUniversityOfCaliforniaLicenseAgreementMember bbio:TheRasIncMember 2020-01-01 2020-12-31 0001743881 bbio:RegentsOfUniversityOfCaliforniaMember bbio:TheRegentsOfTheUniversityOfCaliforniaLicenseAgreementMember bbio:TheRasIncMember 2019-01-01 2019-12-31 0001743881 bbio:RegentsOfUniversityOfCaliforniaMember bbio:TheRegentsOfTheUniversityOfCaliforniaLicenseAgreementMember bbio:TheRasIncMember 2018-01-01 2018-12-31 0001743881 bbio:TheRasIncMember bbio:LeidosBiomedicalResearchLicenseAndCooperativeResearchAndDevelopmentAgreementsMember bbio:LeidosBiomedicalResearchIncMember 2020-01-01 2020-12-31 0001743881 bbio:TheRasIncMember bbio:LeidosBiomedicalResearchLicenseAndCooperativeResearchAndDevelopmentAgreementsMember bbio:LeidosBiomedicalResearchIncMember 2019-01-01 2019-12-31 0001743881 bbio:TheRasIncMember bbio:LeidosBiomedicalResearchLicenseAndCooperativeResearchAndDevelopmentAgreementsMember bbio:LeidosBiomedicalResearchIncMember 2018-01-01 2018-12-31 0001743881 bbio:QEDTherapeuticsIncMember bbio:FoundationMedicineDiagnosticsAgreementMember bbio:FoundationMedicineIncMember 2020-01-01 2020-12-31 0001743881 bbio:QEDTherapeuticsIncMember bbio:FoundationMedicineDiagnosticsAgreementMember bbio:FoundationMedicineIncMember 2019-01-01 2019-12-31 0001743881 bbio:QEDTherapeuticsIncMember bbio:FoundationMedicineDiagnosticsAgreementMember bbio:FoundationMedicineIncMember 2018-01-01 2018-12-31 0001743881 bbio:LaboratoryFacilityMember 2020-12-31 0001743881 bbio:LaboratoryFacilityMember srt:MinimumMember 2020-12-31 0001743881 bbio:LaboratoryFacilityMember srt:MaximumMember 2020-12-31 0001743881 bbio:ManufacturingAgreementMember 2019-12-01 2019-12-31 0001743881 srt:SubsidiariesMember bbio:SeriesDPreferredUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember bbio:SeriesCPreferredUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember bbio:SeriesBPreferredUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember bbio:SeriesAPreferredUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember bbio:FounderUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember bbio:CommonUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember bbio:ManagementIncentiveUnitsMember 2019-06-13 2019-06-13 0001743881 srt:SubsidiariesMember 2019-06-13 2019-06-13 0001743881 us-gaap:IPOMember us-gaap:CommonStockMember 2019-07-01 2019-07-01 0001743881 us-gaap:OverAllotmentOptionMember us-gaap:CommonStockMember 2019-07-01 2019-07-01 0001743881 us-gaap:IPOMember us-gaap:CommonStockMember 2019-07-01 0001743881 us-gaap:CommonStockMember 2019-07-01 2019-07-01 0001743881 us-gaap:CommonStockMember 2019-07-01 0001743881 us-gaap:CommonStockMember bbio:AtTheMarketOfferingMember srt:MaximumMember 2020-07-07 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:BridgeBioServicesIncMember 2020-01-01 2020-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:EidosTherapeuticsIncMember 2020-01-01 2020-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:OtherConsolidatedEntitiesMember 2020-01-01 2020-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember 2020-01-01 2020-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:BridgeBioServicesIncMember 2020-01-01 2020-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:EidosTherapeuticsIncMember 2020-01-01 2020-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:OtherConsolidatedEntitiesMember 2020-01-01 2020-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember 2020-01-01 2020-12-31 0001743881 bbio:BridgeBioServicesIncMember 2020-01-01 2020-12-31 0001743881 bbio:OtherConsolidatedEntitiesMember 2020-01-01 2020-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:BridgeBioServicesIncMember 2019-01-01 2019-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:EidosTherapeuticsIncMember 2019-01-01 2019-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:OtherConsolidatedEntitiesMember 2019-01-01 2019-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember 2019-01-01 2019-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:BridgeBioServicesIncMember 2019-01-01 2019-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:EidosTherapeuticsIncMember 2019-01-01 2019-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:OtherConsolidatedEntitiesMember 2019-01-01 2019-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember 2019-01-01 2019-12-31 0001743881 bbio:BridgeBioServicesIncMember 2019-01-01 2019-12-31 0001743881 bbio:EidosTherapeuticsIncMember 2019-01-01 2019-12-31 0001743881 bbio:OtherConsolidatedEntitiesMember 2019-01-01 2019-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:EidosTherapeuticsIncMember 2018-01-01 2018-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember bbio:OtherConsolidatedEntitiesMember 2018-01-01 2018-12-31 0001743881 us-gaap:ResearchAndDevelopmentExpenseMember 2018-01-01 2018-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:BridgeBioServicesIncMember 2018-01-01 2018-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:EidosTherapeuticsIncMember 2018-01-01 2018-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember bbio:OtherConsolidatedEntitiesMember 2018-01-01 2018-12-31 0001743881 us-gaap:GeneralAndAdministrativeExpenseMember 2018-01-01 2018-12-31 0001743881 bbio:BridgeBioServicesIncMember 2018-01-01 2018-12-31 0001743881 bbio:EidosTherapeuticsIncMember 2018-01-01 2018-12-31 0001743881 bbio:OtherConsolidatedEntitiesMember 2018-01-01 2018-12-31 0001743881 us-gaap:CommonStockMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember 2019-06-25 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember 2020-06-02 2020-06-02 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember 2019-06-24 2019-06-25 0001743881 us-gaap:CommonStockMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember 2019-11-13 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember 2020-12-31 0001743881 bbio:TwoThousandNineteenInducementEquityPlansMember 2020-12-31 bbio:Grantee 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember 2020-04-22 2020-04-22 0001743881 us-gaap:EmployeeStockOptionMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-04-22 2020-04-22 0001743881 us-gaap:RestrictedStockMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-04-22 2020-04-22 0001743881 bbio:PerformanceBasedRestrictedStockAwardsMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-04-22 2020-04-22 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-04-22 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-11-18 2020-11-18 0001743881 us-gaap:EmployeeStockOptionMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-11-18 2020-11-18 0001743881 bbio:PerformanceBasedStockOptionsMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-11-18 2020-11-18 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-11-18 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-12-31 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember srt:MinimumMember 2020-04-22 2020-04-22 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember srt:MinimumMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember srt:MaximumMember 2020-04-22 2020-04-22 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember srt:MaximumMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2019-01-01 2019-12-31 0001743881 bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember us-gaap:RestrictedStockMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember 2019-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember 2018-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlansMember 2019-01-01 2019-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:EmployeeStockOptionMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:EmployeeStockOptionMember 2019-01-01 2019-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:EmployeeStockOptionMember 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:RestrictedStockUnitsRSUMember 2019-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:RestrictedStockUnitsRSUMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:RestrictedStockUnitsRSUMember 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:RestrictedStockUnitsRSUMember 2019-01-01 2019-12-31 0001743881 us-gaap:RestrictedStockMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember 2019-12-31 0001743881 us-gaap:RestrictedStockMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember 2020-01-01 2020-12-31 0001743881 us-gaap:RestrictedStockMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember 2020-12-31 0001743881 us-gaap:RestrictedStockMember bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember bbio:TwoThousandTwentyStockAndEquityAwardExchangeProgramMember 2020-01-01 2020-12-31 0001743881 bbio:TwoThousandNineteenStockOptionAndIncentivePlanAndInducementEquityPlanMember us-gaap:RestrictedStockMember 2019-01-01 2019-12-31 0001743881 bbio:MarketBasedRestrictedStockUnitsMember 2019-01-01 2019-12-31 0001743881 bbio:MarketBasedRestrictedStockUnitsMember 2019-12-31 0001743881 bbio:MarketBasedRestrictedStockUnitsMember 2020-12-31 0001743881 us-gaap:EmployeeStockMember srt:MaximumMember 2019-06-22 0001743881 us-gaap:EmployeeStockMember 2019-06-22 2019-06-22 0001743881 us-gaap:EmployeeStockMember 2019-06-22 0001743881 us-gaap:EmployeeStockMember 2020-01-01 2020-12-31 0001743881 us-gaap:EmployeeStockMember 2019-01-01 2019-12-31 0001743881 us-gaap:EmployeeStockMember 2020-12-31 0001743881 srt:MinimumMember us-gaap:EmployeeStockOptionMember 2020-01-01 2020-12-31 0001743881 us-gaap:EmployeeStockMember srt:MinimumMember 2020-01-01 2020-12-31 0001743881 srt:MinimumMember us-gaap:EmployeeStockOptionMember 2019-01-01 2019-12-31 0001743881 srt:MaximumMember us-gaap:EmployeeStockOptionMember 2020-01-01 2020-12-31 0001743881 us-gaap:EmployeeStockMember srt:MaximumMember 2020-01-01 2020-12-31 0001743881 srt:MaximumMember us-gaap:EmployeeStockOptionMember 2019-01-01 2019-12-31 0001743881 us-gaap:EmployeeStockOptionMember 2020-01-01 2020-12-31 0001743881 us-gaap:EmployeeStockOptionMember 2019-01-01 2019-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2020-01-01 2020-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember srt:MinimumMember 2020-01-01 2020-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember srt:MaximumMember 2020-01-01 2020-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2019-01-01 2019-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2018-01-01 2018-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2017-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2018-01-01 2018-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2018-12-31 0001743881 bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2019-01-01 2019-12-31 0001743881 srt:MinimumMember bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2018-01-01 2018-12-31 0001743881 srt:MaximumMember bbio:ManagementIncentiveUnitsAndCommonUnitsMember srt:SubsidiariesMember 2018-01-01 2018-12-31 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanMember srt:MinimumMember bbio:IncentiveStockOptionsAndNonqualifiedStockOptionsMember 2016-04-01 2016-04-30 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanMember bbio:IncentiveStockOptionsMember 2016-04-01 2016-04-30 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanMember srt:MinimumMember bbio:IncentiveStockOptionsMember 2016-04-01 2016-04-30 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanMember bbio:IncentiveStockOptionsAndNonqualifiedStockOptionsMember 2016-04-01 2016-04-30 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanMember 2020-01-01 2020-12-31 0001743881 bbio:AmendedAndRestatedTwoThousandEighteenStockOptionAndIncentivePlanMember 2018-05-31 0001743881 bbio:AmendedAndRestatedTwoThousandEighteenStockOptionAndIncentivePlanMember 2018-05-01 2018-05-31 0001743881 bbio:AmendedAndRestatedTwoThousandEighteenStockOptionAndIncentivePlanMember srt:MinimumMember 2018-05-01 2018-05-31 0001743881 bbio:AmendedAndRestatedTwoThousandEighteenStockOptionAndIncentivePlanMember 2018-12-01 2018-12-31 0001743881 bbio:AmendedAndRestatedTwoThousandEighteenStockOptionAndIncentivePlanMember 2019-12-01 2019-12-31 0001743881 bbio:AmendedAndRestatedTwoThousandEighteenStockOptionAndIncentivePlanMember 2020-12-31 0001743881 bbio:TwoThousandEighteenEmployeeStockPurchasePlanMember 2018-05-31 0001743881 bbio:TwoThousandEighteenEmployeeStockPurchasePlanMember 2018-05-01 2018-05-31 0001743881 bbio:TwoThousandEighteenEmployeeStockPurchasePlanMember 2019-01-01 2019-12-31 0001743881 bbio:TwoThousandEighteenEmployeeStockPurchasePlanMember 2018-01-01 2018-12-31 0001743881 bbio:EidosTherapeuticsIncMember 2019-12-31 0001743881 bbio:EidosTherapeuticsIncMember 2020-12-31 0001743881 bbio:EmployeeMember bbio:EidosTherapeuticsIncMember 2019-01-01 2019-12-31 0001743881 bbio:EmployeeMember bbio:EidosTherapeuticsIncMember 2018-01-01 2018-12-31 0001743881 us-gaap:RestrictedStockMember 2017-12-01 2017-12-31 0001743881 us-gaap:RestrictedStockMember us-gaap:ShareBasedCompensationAwardTrancheOneMember 2017-12-01 2017-12-31 0001743881 us-gaap:RestrictedStockMember us-gaap:ShareBasedCompensationAwardTrancheTwoMember 2017-12-01 2017-12-31 0001743881 us-gaap:RestrictedStockMember 2020-12-31 0001743881 us-gaap:RestrictedStockMember 2019-12-31 0001743881 us-gaap:RestrictedStockMember 2020-01-01 2020-12-31 0001743881 us-gaap:RestrictedStockMember 2019-01-01 2019-12-31 0001743881 us-gaap:RestrictedStockMember 2018-01-01 2018-12-31 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanAndTwoThousandEighteenStockOptionAndIncentivePlanMember 2020-12-31 0001743881 bbio:TwoThousandSixteenEquityIncentivePlanAndTwoThousandEighteenStockOptionAndIncentivePlanMember 2020-01-01 2020-12-31 0001743881 us-gaap:DomesticCountryMember 2020-12-31 0001743881 us-gaap:StateAndLocalJurisdictionMember country:CA 2020-12-31 0001743881 2017-01-01 2017-12-31 0001743881 us-gaap:DomesticCountryMember bbio:ResearchAndDevelopmentAndOrphanDrugCreditCarryforwardsMember 2020-12-31 0001743881 us-gaap:StateAndLocalJurisdictionMember us-gaap:ResearchMember 2020-12-31 0001743881 us-gaap:DomesticCountryMember bbio:ResearchAndDevelopmentAndOrphanDrugCreditCarryforwardsMember 2020-01-01 2020-12-31 0001743881 bbio:UnvestedRestrictedStockAwardMember 2020-01-01 2020-12-31 0001743881 bbio:UnvestedRestrictedStockAwardMember 2019-01-01 2019-12-31 0001743881 bbio:UnvestedRestrictedStockAwardMember 2018-01-01 2018-12-31 0001743881 bbio:UnvestedRestrictedStockUnitMember 2020-01-01 2020-12-31 0001743881 bbio:UnvestedRestrictedStockUnitMember 2019-01-01 2019-12-31 0001743881 bbio:UnvestedMarketBasedRestrictedStockUnitMember 2020-01-01 2020-12-31 0001743881 bbio:UnvestedMarketBasedRestrictedStockUnitMember 2019-01-01 2019-12-31 0001743881 bbio:UnvestedPerformanceBasedRestrictedStockUnitMember 2020-01-01 2020-12-31 0001743881 bbio:UnvestedPerformanceBasedRestrictedStockAwardMember 2020-01-01 2020-12-31 0001743881 bbio:CommonStockOptionsIssuedAndOutstandingMember 2020-01-01 2020-12-31 0001743881 bbio:CommonStockOptionsIssuedAndOutstandingMember 2019-01-01 2019-12-31 0001743881 bbio:EstimatedSharesIssuablePerformanceBasedMilestoneCompensationArrangementMember 2020-01-01 2020-12-31 0001743881 bbio:EstimatedSharesIssuableEmployeeStockPurchasePlanMember 2020-01-01 2020-12-31 0001743881 bbio:AssumedConversionOfTwoThousandTwentySevenNotesMember 2020-01-01 2020-12-31 0001743881 us-gaap:SubsequentEventMember bbio:EidosTherapeuticsIncMember 2021-01-26 2021-01-26 0001743881 us-gaap:SubsequentEventMember bbio:EidosTherapeuticsIncMember bbio:TwoThousandSixteenEquityIncentivePlanAndTwoThousandEighteenStockOptionAndIncentivePlanMember 2021-01-26 2021-01-26 0001743881 us-gaap:OtherCurrentAssetsMember bbio:EidosTherapeuticsIncMember 2020-12-31 0001743881 us-gaap:SubsequentEventMember bbio:EidosTherapeuticsIncMember 2021-01-26 0001743881 bbio:TwoPointTwoFivePercentageConvertibleSeniorNotesDueTwoThousandAndTwentyNineMember us-gaap:SubsequentEventMember 2021-01-28 0001743881 bbio:TwoPointTwoFivePercentageConvertibleSeniorNotesDueTwoThousandAndTwentyNineMember us-gaap:SubsequentEventMember 2021-01-28 2021-01-28

 

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, 2020

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 No. 001-38959

BridgeBio Pharma, Inc.

(Exact name of registrant as specified in its charter)

 

Delaware
(State or other jurisdiction of
incorporation or organization)

84-1850815
(I.R.S. Employer
Identification No.)

421 Kipling Street, Palo Alto, CA
(Address of principal executive offices)

94301
(Zip Code)

 

Registrant’s telephone number, including area code: (650) 391-9740

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, par value $0.001 per share

 

BBIO

 

The 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 Section 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. 726(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 voting and non‑voting common equity held by non‑affiliates of the registrant based upon the closing price of the registrant’s Common Stock on The Nasdaq Global Select Market on June 30, 2020 was approximately $1,694.0 million. Shares of the registrant’s Common Stock held by each executive officer and director and by each other person who may be deemed an affiliate of the Registrant have been excluded from this computation. The determination of affiliate status for this purpose is not necessarily a conclusive determination for other purposes.

On February 19, 2021, there were 148,956,329 shares of the registrant’s Common Stock issued and outstanding.

DOCUMENTS INCORPORATED BY REFERENCE

Specified portions of the registrant’s definitive Proxy Statement to be issued in conjunction with the registrant’s 2021 Annual Meeting of Stockholders, which is expected to be filed not later than 120 days after the registrant’s fiscal year ended December 31, 2020, are incorporated by reference into Part III of this Annual Report. Except as expressly incorporated by reference, the registrant’s Proxy Statement shall not be deemed to be a part of this Annual Report on Form 10‑K.

 

 

 

 


BRIDGEBIO PHARMA, INC.

2020 Form 10‑K Annual Report

Table of Contents

 

 

 

Page

 

PART I

 

Item 1.

Business

1

Item 1A.

Risk Factors

52

Item 1B.

Unresolved Staff Comments

115

Item 2.

Properties

115

Item 3.

Legal Proceedings

115

Item 4.

Mine Safety Disclosures

115

 

PART II

 

Item 5.

Market for the Registrant’s Common Equity, Related Stockholder Matters and Issuer Purchases of Equity Securities

116

Item 6.

Selected Financial Data

117

Item 7.

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

118

Item 7A.

Quantitative and Qualitative Disclosures About Market Risk

134

Item 8.

Financial Statements and Supplementary Data

135

Item 9.

Changes in and Disagreements with Accountants on Accounting and Financial Disclosure

195

Item 9A.

Controls and Procedures

195

Item 9B.

Other Information

196

 

PART III

 

Item 10.

Directors, Executive Officers and Corporate Governance

197

Item 11.

Executive Compensation

197

Item 12.

Security Ownership of Certain Beneficial Owners and Management and Related Stockholder Matters

197

Item 13.

Certain Relationships and Related Transactions, and Director Independence

197

Item 14.

Principal Accountant Fees and Services

197

 

PART IV

 

Item 15.

Exhibits and Financial Statement Schedules

198

Item 16.

Form 10‑K Summary

199

Exhibits

199

Signatures

205

 

i


Special Note Regarding Forward‑Looking Statements

This Annual Report on Form 10‑K contains forward‑looking statements within the meaning of Section 27A of the Securities Act of 1933, as amended, and Section 21E of the Securities Exchange Act of 1934, as amended. Such forward‑looking statements involve substantial risks, uncertainties and assumptions. All statements in this Annual Report on Form 10‑K, other than statements of historical fact, including, without limitation, statements regarding our strategy, future operations, future financial position, future revenue, projected costs, prospects, plans, intentions, expectations, goals and objectives may be forward‑looking statements. The words “anticipates,” “believes,” “could,” “designed,” “estimates,” “expects,” “goal,” “intends,” “may,” “objective,” “plans,” “projects,” “pursuing,” “will,” “would” and similar expressions (including the negatives thereof) are intended to identify forward‑looking statements, although not all forward‑looking statements contain these identifying words. The forward-looking statements in this report include, but are not limited to, statements about:

 

the extent to which the coronavirus disease 2019, or COVID-19, pandemic and measures taken to contain its spread ultimately impact our business, including our clinical development and research activities;

 

the success, cost and timing of our clinical development of our product candidates, including the progress of, and results from, our ongoing and planned Phase 3 clinical trials of acoramidis, our ongoing and planned Phase 2 and Phase 3 clinical trials of infigratinib, our planned clinical trial of BBP-631 and our ongoing Phase 2 clinical trial of encaleret, as well as the potential indications for each;

 

 

potential adverse effects or changes to relationships with customers, employees, suppliers or other parties resulting from the completion of our acquisition of the shares of Eidos Therapeutics, Inc., or Eidos, common stock that we did not already own, or the Eidos Merger;

 

 

pending and potential future complaints and legal proceedings relating to the Eidos Merger that have been and could be instituted against us, Eidos or our or their respective directors and officers, including the effects of any outcomes related thereto;

 

 

possible disruptions from the Eidos Merger that could harm our or Eidos’ business, including current plans and operations;

 

 

unexpected costs, charges or expenses resulting from the Eidos Merger;

 

 

uncertainty of our and Eidos’ expected financial performance following completion of the Eidos Merger, including the possibility that the expected synergies and value creation from the merger will not be realized or will not be realized within the expected time period;

 

 

our and Eidos’ ability to implement our respective business strategies following the Eidos Merger;

 

 

our and Eidos’ ability to continue planned preclinical and clinical development of our respective development programs, and the timing, cost and success of any such continued preclinical and clinical development and planned regulatory submissions;

 

our ability to initiate, recruit and enroll patients in and conduct our clinical trials at the pace that we project;

 

the timing of our submissions to the U.S. Food and Drug Administration and any review or comments on data that we will need to generate to file our Investigational New Drug applications or New Drug Applications, including pending or new clinical hold notices;

 

our plans to implement certain development strategies, including our ability to attract and retain potential collaborators with development, regulatory and commercialization expertise;

 

our ability to obtain and maintain regulatory approval of our product candidates in any of the indications for which we plan to develop them, and any related restrictions, limitations or warnings in the label of any of our product candidates, if approved;

ii


 

our ability to compete with companies currently marketing approved treatments or engaged in the development of treatments that may become available for any of the indications that our product candidates are designed to target;

 

our reliance on third parties to conduct our clinical trials and to manufacture drug substance for use in our clinical trials;

 

our ability to contract with and the performance of our and our collaborators’ third-party suppliers and manufacturers;

 

the size and growth potential of the markets for acoramidis, low-dose infigratinib, BBP-631, encaleret and any of our current product candidates or other product candidates we may identify and pursue, and our ability to serve and our acceptance by those markets;

 

our ability to identify and advance through clinical development any additional product candidates;

 

the commercialization of our current product candidates and any other product candidates we may identify and pursue, if approved, including our ability to successfully build a specialty sales force and commercial infrastructure to market our current product candidates and any other product candidates we may identify and pursue;

 

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

 

our ability to obtain and maintain adequate intellectual property rights for our product candidates and our ability to operate our business without infringing on the intellectual property rights of others;

 

our expectations regarding government and third-party payor coverage and reimbursement;

 

our estimates of our expenses, ongoing losses, capital requirements and our use of cash resources, and our needs for or ability to obtain additional financing to complete the clinical trials of any of our product candidates;

 

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

 

our financial performance; and

 

developments and projections relating to our competitors or our industry.

We may not actually achieve the plans, intentions, expectations or objectives disclosed in our forward‑looking statements and the assumptions underlying our forward‑looking statements may prove incorrect. Furthermore, if our forward‑looking statements prove to be inaccurate, the inaccuracy may be material. Therefore, you should not place undue reliance on our forward‑looking statements, and you should not regard these statements as a representation or warranty by us or any other person that we will achieve our objectives and plans in any specified time frame, or at all. Actual results or events could differ materially from the plans, intentions, expectations and objectives disclosed in the forward‑looking statements that we make. Important factors that we believe could cause actual results or events to differ materially from our forward‑looking statements include, but are not limited to, those listed under “Risk Factors” in Item 1A of Part I, “Management’s Discussion and Analysis of Financial Condition and Results of Operations” in Item 7 of Part II and elsewhere in this Annual Report on Form 10‑K. Our forward‑looking statements in this Annual Report on Form 10‑K are based on current expectations as of the date hereof and we do not assume any obligation to update any forward‑looking statements on account of new information, future events or otherwise, except as required by law.

iii


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” and should be carefully considered, together with other information in this Annual Report on Form 10-K and our other filings with the U.S. Securities and Exchange Commission, or the SEC, before making investment decisions regarding our common stock.

 

 

Our product candidates are in preclinical or clinical development, which is a lengthy and expensive process with uncertain outcomes and the potential for substantial delays. We may not receive regulatory approval to market any of our product candidates in any jurisdictions, which would materially and adversely affect our business, prospects, operating results and financial condition.

 

Our clinical trials may fail to demonstrate substantial evidence of the safety and efficacy of product candidates that we may identify and pursue for their intended uses, which would prevent, delay or limit the scope of regulatory approval and commercialization.

 

Drug development is a highly uncertain undertaking and involves a substantial degree of risk. We have incurred significant losses since our inception and anticipate that we will continue to incur significant losses for the foreseeable future. We have not generated any revenue since inception, which, together with our limited operating history, may make it difficult for you to assess our future viability.

 

We may encounter substantial delays in clinical trials for a variety of reasons, including difficulties in patient enrollment, and we may not be able to conduct or complete clinical trials on the expected timelines, if at all.

 

Results of earlier studies or clinical trials may not be predictive of future clinical trial results, and initial studies or clinical trials may not establish an adequate safety or efficacy profile for our product candidates to justify proceeding to advanced clinical trials or an application for regulatory approval.

 

Our product candidates could be associated with side effects, adverse events or other properties or safety risks, which could delay or halt their clinical development, prevent their regulatory approval, cause us to abandon a product candidate, limit their commercial potential, if approved, or result in other significant negative consequences to our business, prospects, operating results and financial condition.

 

Certain of our product candidates under development for the treatment of patient populations with significant comorbidities that may result in deaths or serious adverse or unacceptable side effects and require us to abandon or limit our clinical development activities.

 

Interim, “top-line,” and preliminary data from our clinical trials that we announce or publish from time to time may change as more data become available, as additional analyses are conducted, or as audit and verification procedures are performed on such preliminary data.

 

Our conduct of clinical trials for product candidates and our plans to commercialize certain product candidates outside the United States could expose us to additional risks and uncertainties, including with respect to our ability to obtain regulatory approvals or comply with applicable laws and regulations outside the United States.

 

Even though we may apply for orphan drug designation for our product candidates, we may not be able to obtain orphan drug marketing exclusivity.

 

Even if we obtain regulatory approval for a product candidate, we will be subject to ongoing regulatory obligations and continued regulatory review, which may result in significant expense to maintain compliance with such obligations or as a result of any penalties to which we may become subject for any failure to comply with such obligations.

 

Certain of our product candidates, including our protein therapeutic and gene therapy product candidates, are novel, complex and difficult to manufacture. We could experience manufacturing problems that delay our development or commercialization activities or otherwise harm our business.

Certain of our product candidates are based on a novel adeno-associated virus, or AAV, gene therapy technology with which there is limited clinical or regulatory experience to date, which makes it difficult to predict the time and cost of product candidate development and subsequently obtaining regulatory approval.

 

We face significant competition from other biotechnology and pharmaceutical companies, and our operating results will suffer if we fail to compete effectively.

iv


 

Our future success depends on our ability to retain key employees, directors, consultants and advisors and to attract, retain and motivate qualified personnel.

 

We will need substantial additional financing to develop our product candidates and implement our operating plans. If we fail to obtain additional financing, we may be unable to complete the development and commercialization of our product candidates.

 

We rely and will continue to rely on third parties to conduct our clinical trials and some aspects of our research and preclinical testing, and for the manufacture of our product candidates for preclinical studies and clinical trials. If these third parties do not successfully carry out their contractual duties or meet expected deadlines, we may not be able to obtain regulatory approval of or commercialize our product candidates.

 

Our rights to develop and commercialize our product candidates are subject in part to the terms and conditions of licenses granted to us by others, and the patent protection, prosecution and enforcement for some of our product candidates may be dependent on our licensors, the loss of which could result in the loss of intellectual property and other protection, and would harm our business.

 

If we are unable to obtain and maintain sufficient intellectual property protection for our product candidates, including acoramidis, low-dose infigratinib, BBP-631 and encaleret, or if the scope of the intellectual property protection obtained is not sufficiently broad, our competitors could develop and commercialize product candidates similar or identical to ours, and our ability to successfully commercialize our product candidates may be impaired.

 

Even if any product candidates we develop receive marketing approval, they may fail to achieve the degree of market acceptance by physicians, patients, healthcare payors, and others in the medical community necessary for commercial success.

 

Because we have multiple programs and product candidates in our development pipeline and are pursuing a variety of target indications and treatment modalities, we may expend our limited resources to pursue a particular product candidate and fail to capitalize on development opportunities or product candidates that may be more profitable or for which there is a greater likelihood of success.

 

We have incurred a significant amount of debt and may in the future incur additional indebtedness. Servicing our debt requires a significant amount of cash, and we may not have sufficient cash flow from our business to pay our substantial debt.

 

There is uncertainty regarding our and Eidos’ expected financial performance following completion of our acquisition, including the possibility that the expected synergies and value creation from the merger following the acquisition will not be realized or will not be realized within the expected time period.

 

The outbreak of the novel strain of coronavirus, SARS-CoV-2, which causes COVID-19, could adversely impact our business, including our clinical trials and preclinical studies.

 

 

 

v


PART I

ITEM 1.  BUSINESS

Overview

We are a team of experienced drug discoverers, developers and innovators working to create life-altering medicines that target well-characterized genetic diseases at their source. We founded BridgeBio in 2015 to identify and advance transformative medicines to treat patients who suffer from Mendelian diseases, which are diseases that arise from defects in a single gene, and cancers with clear genetic drivers. Our pipeline of over 30 development programs includes product candidates ranging from early discovery to late-stage development. We have filed New Drug Applications, or NDAs, with the U.S. Food and Drug Administration, or FDA, for two of our programs. Several of our programs target indications that we believe present the potential for our product candidates, if approved, to target portions of market opportunities of at least $1.0 billion in annual sales.

We focus on genetic diseases because they exist at the intersection of high unmet patient need and tractable biology. Our approach is to translate research pioneered at academic laboratories and leading medical institutions into products that we hope will ultimately reach patients. We are able to realize this opportunity through a confluence of scientific advances, including: (i) identification of the genetic underpinnings of disease as more cost-efficient genome and exome sequencing becomes available; (ii) progress in molecular biology; and (iii) the development and maturation of longitudinal data and retrospective studies that enable the linkage of genes to diseases. We believe that this early-stage innovation represents one of the greatest practical sources for new drug creation.

We believe we have developed a world-class drug engineering product platform that supports the continued growth of our company and the advancement of our pipeline. Leveraging our product platform, we have a goal of adding two to three programs to our pipeline each year going forward.

Our Platform

Our drug engineering platform is characterized by four stages: Discover, Create, Test and Deliver.

 

Discover. We identify novel genetic disease targets and focus only on well-described diseases that can be targeted at their source. Our target identification engine is driven by three core areas of strength. We pair a systematic mapping of the 7,000 known genetic diseases with leading computational genomics and statistical genetics capabilities and partner with top academic institutions. We currently have over 15 academic partnerships. Our target identification process is overseen by an investment committee that is comprised of renowned leaders in cancer and rare disease drug development with proven track records.

 

Create. We select the optimal therapeutic modality to target each disease at its source. We have industry-leading capabilities across four modalities: medicinal chemistry, therapeutic proteins, gene therapy and antisense oligonucleotides. We choose the treatment modality that we believe is best biologically suited to address the root cause of the disease. Our research leaders have a productive history of developing novel therapeutics. Together with the investment committee, the team has previously submitted over 100 Investigational New Drug applications, or INDs, and 20 NDAs, in aggregate.

 

Test. We have built a leading, global clinical development footprint. We have over 18 ongoing clinical trials across five different therapeutic areas, at over 400 trial sites in 26 countries. Across these trials, we have built a central information repository and operational toolkit for best practices for enrollment, protocol quality, site activation, contract research organization, or CRO, quality and regional performance. Our development is led by expert, dedicated teams in each therapeutic area who have the expertise to pursue unique and creative clinical and regulatory strategy.

 

Deliver. We are building capabilities to deliver our potential products to patients across the globe. We intend to build global commercial infrastructure to leverage our drug and disease expertise, including diagnostic partnerships, disease awareness strategies and country-specific early access programs. We have already pursued commercial partners in strategic geographies for select programs.

1


We believe we have developed a world class drug engineering platform that is sustainable and scalable, and already producing results. The discovery platform has yielded over 30 programs in the pipeline. We have filed over 10 INDs since 2015 and are testing our product candidates in over 18 clinical trials across the globe.

Our Pipeline

Our pipeline includes over 30 development programs. The following table summarizes our development programs, their estimated patient populations, their therapeutic modalities and their development status:

 

Of our development programs, we believe the following, to which we refer as our core value drivers, have the greatest potential to drive significant near-term value for our company due to a combination of factors, including their stage of development, potential availability of expedited development pathways, degree of unmet medical need and potential market size in the applicable target indication:

 

Acoramidis (also known as AG10 and BBP-265), a small molecule stabilizer of transthyretin, or TTR, that is in an ongoing Phase 3 clinical trial for the treatment of TTR amyloidosis-cardiomyopathy, or ATTR-CM.

 

Low-dose infigratinib (also known as BBP-831), a small molecule selective FGFR1-3 inhibitor that is an ongoing Phase 2 clinical trial for the treatment of achondroplasia in pediatric patients.

 

BBP-631, an AAV5 gene transfer product candidate for the treatment of congenital adrenal hyperplasia, or CAH, driven by 21-hydroxylase deficiency, or 21OHD, for which we anticipate initiating a first-in-human Phase 1/2 trial in the second half of 2021, with initial data anticipated in late 2021 or early 2022.

 

Encaleret, a small molecule antagonist of the calcium sensing receptor, or CaSR, that is in an ongoing Phase 2 proof-of-concept clinical trial for Autosomal Dominant Hypocalcemia Type 1, or ADH1.

 

2


CORE VALUE DRIVERS

Acoramidis (Eidos): TTR Amyloidosis

Summary

We are developing acoramidis, also known as AG10 and BBP-265, an oral small molecule TTR, stabilizer, for the treatment of TTR amyloidosis, or ATTR. A Phase 3 clinical trial in patients with ATTR-CM, known as the ATTRibute-CM study, is currently ongoing. We anticipate reporting top-line 12-month 6-minute walking distance, or 6MWD, data from the ATTRibute-CM study in late 2021 or early 2022.

Disease Overview

ATTR is a disease caused by destabilization of TTR tetramers resulting in progressive amyloid deposition. TTR is a protein that occurs naturally in the form of a tetramer, which is a molecular structure consisting of four identical subunits, or monomers, and performs multiple physiologic roles, including the transport of essential hormones and vitamins. In ATTR, TTR tetramers become destabilized due to a mutation in the TTR gene or as part of the natural aging process. Destabilized TTR dissociates into monomers, self-aggregates and assembles into fibrils which are deposited, predominantly in the heart and nervous system, driving disease pathophysiology.

ATTR is commonly categorized by its genotypic cause and primary clinical manifestation: wild-type ATTR cardiomyopathy, or ATTRwt-CM, which results from an age-related process; variant ATTR cardiomyopathy, or ATTRv-CM; and ATTR polyneuropathy, or ATTR-PN, which is only associated with TTR variants. All three forms of the disease are progressive and fatal. ATTRwt-CM and ATTRv-CM patients generally present with symptoms later in life (older than 50) and have median life expectancies of three to five years from diagnosis if untreated. ATTR-PN presents either in a patient’s early 30s or later (older than 50), and results in a median life expectancy of five to ten years from diagnosis. Progression of all forms of the disease causes significant disability, impacts productivity and quality of life, and creates a significant economic burden due to the costs associated with patient need for supportive care. As the disease progresses, ATTRwt-CM and ATTRv-CM patients may require frequent hospitalizations and repeated interventions. ATTR-PN patients experience gradual loss of the ability to walk without assistance, and autonomic nervous system function affecting digestion and blood pressure.

The worldwide estimated prevalence of ATTRwt-CM, ATTRv-CM, and ATTR-PN is approximately 400,000, 40,000, and 10,000, respectively. However, we believe that the cardiomyopathic forms of the disease are significantly underdiagnosed. For example, recent literature has suggested that between 12% to 19% of patients diagnosed with heart failure with preserved ejection fraction may, in fact, have undiagnosed ATTR-CM. This single segment represents approximately half of the 6.0 million to 7.0 million estimated people with heart failure in the United States alone. With the increasing availability of disease-modifying therapeutics, disease awareness is heightened.

We believe the population of diagnosed ATTR-CM patients is also growing rapidly due to the shift to an accurate and reliable non-invasive diagnostic imaging technique. Historically, a heart biopsy was required to make a diagnosis of ATTR-CM. Recently, however, it has been shown that scintigraphy with technetium-labelled radiotracers is a highly accurate, non-invasive, and cost-effective method for ATTR-CM diagnosis. We believe that both increased disease awareness and availability of this non-invasive diagnostic imaging technique are allowing for earlier diagnosis of ATTR-CM patients and the identification of previously misdiagnosed patients.

Our Product Concept

Acoramidis is a clinical-stage, orally administered, small molecule TTR stabilizer being developed to treat ATTR at its source by reducing the level of amyloid formation through TTR stabilization. This has been shown in preclinical studies and clinical trials to prevent the dissociation of tetrameric TTR into monomers, and in preclinical studies, to reduce the rate of amyloid fibril formation. In addition, acoramidis has been shown to lead to increased circulating levels of tetrameric TTR. Acoramidis has been designed to bind TTR in a way that causes TTR’s conformational structure to mimic that of the well-characterized T119M variant, a naturally occurring rescue mutation that super stabilizes the TTR tetramer. The T119M variant has been observed to prevent the dissociation of

3


TTR tetramers into monomers; T119M tetramers dissociate 40-fold more slowly than wild-type tetramers in biochemical assays. Known as a trans-allelic trans-suppressor, individuals who coinherit the T119M rescue mutation along with a TTR-destabilizing mutation are protected against the development of ATTR.

In third-party clinical trials of tafamidis and diflunisal, interventional approaches that increased TTR stabilization led to improved outcomes in this disease and were correlated with increases in serum TTR. Further, based on genetic data, there is a correlation between the level of TTR stabilization, serum TTR levels and disease severity. As a result, we believe that serum TTR is a predictive biomarker for disease prognosis and that there may be a relationship between more effective TTR stabilization, serum TTR levels and improved clinical outcomes. Based on head-to-head preclinical data, we believe that acoramidis has the potential to stabilize TTR to a greater extent than other TTR stabilizers.

Clinical Data

Phase 2 Data

In April 2018, we initiated our Phase 2 randomized, placebo-controlled, dose-ranging clinical trial of acoramidis in 49 patients with symptomatic ATTR-CM, of which 14 had ATTRv-CM. Eligible patients were randomized in a 1:1:1 ratio to placebo or 400 mg or 800 mg of acoramidis twice daily. The primary objective of the trial was to evaluate the safety and tolerability of acoramidis administered to adult subjects with symptomatic ATTR-CM. The secondary objectives were to characterize the pharmacokinetics, or PK, of acoramidis in symptomatic ATTR-CM subjects and to describe the pharmacodynamics, or PD, properties of acoramidis, as well as the PK-PD relationship of acoramidis. The PD assessments of TTR stabilization were measured by fluorescent probe exclusion, Western blot and serum prealbumin (TTR). The trial design is depicted below:

 

 

4


Enrolled symptomatic ATTR-CM subjects ranged in age from 60 to 86 years of age, with a mean age of 74.1, and 92% of subjects were male. In this trial, we enrolled subjects exclusively with advanced disease, with 29% of subjects presenting with New York Heart Association, or NYHA, Class III heart failure symptoms and a high baseline NT-proBNP with a mean of 3,368 pg/mL. Additionally, on average, subjects had relatively low TTR at baseline with a mean of 22.0 mg/dL. The laboratory reference range for serum TTR is 20 mg/dL to 40 mg/dL in healthy individuals. Both high NT-proBNP and low TTR are biomarkers of disease severity. The subject disposition and baseline characteristics are shown below.

 

 

 

Placebo

 

Acoramidis 400 mg

 

Acoramidis 800 mg

 

Total

Characteristic

 

(n = 17)

 

(n = 16)

 

(n = 16)

 

(n = 49)

Age, mean (range)

 

73.2 (60-5)

 

73.8 (60-83)

 

75.4 (67-86)

 

74.1 (60-86)

Male, n (%)

 

17 (100%)

 

14 (88%)

 

14 (88%)

 

45 (92%)

ATTRv-CM, n (%)

 

3 (18%)

 

6 (38%)

 

5 (31%)

 

14 (29%)

NYHA Class III, n (%)

 

5 (29%)

 

6 (38%)

 

3 (19%)

 

14 (29%)

Race, n (%)

 

 

 

 

 

 

 

 

White

 

13 (76%)

 

10 (62%)

 

12 (75%)

 

35 (72%)

Black

 

3 (18%)

 

4 (25%)

 

3 (19%)

 

10 (20%)

Other

 

1 (6%)

 

2 (13%)

 

1 (6%)

 

4 (8%)

NT-proBNP (pg/mL)1

 

3151 ± 2705

 

3589 ± 3020

 

3377 ± 2806

 

3368 ± 2789

Troponin I (ng/mL)2

 

0.17 ± 0.30

 

0.22 ± 0.24

 

0.10 ± 0.06

 

0.16 ± 0.22

TTR (mg/dL)3

 

23.4 ± 5.5

 

23.2 ± 5.7

 

19.5 ± 4.2

 

22.0 ± 5.4

 

1

NT-proBNP normal range = 0 – 449 pg/mL; NT-proBNP = N-Terminal pro B-type Natriuretic Peptide

2

Troponin I normal range = 0 – 0.02 ng/mL

3

TTR normal range = 20 – 40 mg/dL

Overall, acoramidis was well-tolerated in symptomatic ATTR-CM subjects with no lab safety signals of potential clinical concern attributed to study drug. In this trial, 88% of subjects administered placebo experienced adverse events, or AEs, and 63% and 69% of subjects administered 400 mg and 800 mg acoramidis experienced AEs, respectively. In both the placebo and active treatment groups, most of the AEs were mild to moderate in severity. The most commonly observed AEs, occurring in four or more subjects across the treatment and placebo groups, were atrial fibrillation, constipation, diarrhea and muscle spasms. Three subjects reported serious adverse events, or SAEs, during this study. One placebo-treated subject experienced two SAEs, of atrial fibrillation and congestive heart failure and another placebo-treated subject experienced cellulitis in their lower extremity. One acoramidis treated subject experienced an SAE of shortness of breath on study, which was considered unlikely to be related to study drug.

As shown in the chart below, subjects in the placebo group experienced a mean 7% reduction in the circulating tetrameric TTR concentrations compared to baseline. Conversely, subjects administered either 400 mg or 800 mg acoramidis showed a dose-dependent statistically significant mean increase in circulating TTR of 36% and 50%, respectively, compared to baseline. Compared to placebo, both the 400 mg and 800 mg acoramidis arms demonstrated statistically significant increases in mean circulating TTR (p<0.0001 for both arms). p-value is a statistical calculation that relates to the probability that the difference between groups happened by chance, with a p-value of less than 0.05 (i.e., less than 5% probability that the difference happened by chance) generally being used as the threshold to indicate statistical significance. There was a greater observed treatment effect in subjects with variant ATTR-CM as compared to subjects with wild-type ATTR-CM, which we believe can be explained, in part, by the lower absolute serum TTR of variant ATTR-CM subjects at baseline.

 

 

5


 

 

The following chart shows that treatment with acoramidis restored serum TTR concentrations to within the normal range in all subjects at Day 28.

 

 

6


Ex vivo stabilization assays demonstrated near-complete TTR stabilization by acoramidis, with greater than 90% average tetramer stabilization across subjects treated with 400 mg and 800 mg acoramidis, as shown in the chart below. The stabilization response was consistent across variant and wild-type TTR carriers and replicates previously reported clinical and preclinical TTR stabilization data.

 

 

 

Interim analysis of the ongoing Phase 2 open-label extension, or OLE, study was completed on August 31, 2019 in conjunction with annual regulatory reporting and review, at which time 41 participants remained in the study. Three (6.4%) participants in the OLE had died, two due to disease progression and one due to cervical cancer. Three (6.4%) additional patients enrolled in the study had discontinued treatment, including one participant who underwent cardiac transplantation for their disease.

AEs reported in the OLE study were generally consistent with the underlying ATTR-CM disease state and no safety signals of potential clinical concern were associated with the administration of acoramidis in the study. Forty-six (97.9%) patients experienced a treatment-emergent AE reported during the study, with falls, congestive cardiac failure, dyspnea and acute kidney injury the most commonly reported AEs. Nineteen (40.4%) participants experienced a treatment-emergent SAE reported during the study, with congestive cardiac failure (10.6%) and acute kidney injury (8.5%) the most commonly reported SAEs.

The rate of all-cause mortality (including either death or cardiac transplantation, 8.5%) and cardiovascular-related hospitalizations (25.5%) observed in an exploratory analysis of participants in this study following a median of 15 months since Phase 2 initiation were lower than those observed at 15 months in placebo-treated patients in the ATTR-ACT study (all-cause mortality including death or cardiac transplantation, 15.3%; cardiovascular-related hospitalizations, 41.8%).

7


Stabilization of TTR, as measured using established ex vivo assays, was maintained greater than 90% on average at all study visits in actively treated patients.

 

 

(1) Reported occupancy >100% caused by background protein fluorescence.

 

Mean serum TTR levels, a prognostic indicator of survival in a published cohort of wild-type ATTR-CM patients, were elevated upon acoramidis treatment and were maintained in the normal range throughout the study duration. Mean serum TTR levels were increased from baseline by 39% and 56% in wild-type and variant-carrying ATTR-CM patients, respectively, at OLE Visit Day 180.

 

 

1

400mg and 800mg BID acoramidis groups pooled during randomized portion

8


2

Defined as the lower limit of the reference interval for the serum prealbumin (TTR) clinical laboratory assay

Cardiac biomarkers and echocardiographic parameters were stable during the OLE study. NT-proBNP and TnI were unchanged throughout the course of the study. Echocardiographic parameters, including left ventricular mass and left ventricular stroke volume index, were unchanged during the study.

Clinical Development Plan

We are conducting a randomized, global Phase 3 study of acoramidis in ATTR-CM patients, or ATTRibute-CM. ATTRibute-CM enrolled 632 subjects with symptomatic ATTR-CM, including both wild-type and variant TTR carriers with NYHA Class I-III symptoms. Subjects were randomized 2:1 between treatment (acoramidis 800 mg twice daily) and placebo. In Part A, change in 6MWD at 12 months will be compared between treatment and placebo groups as a potential registrational endpoint. We anticipate reporting 12-month 6MWD data from Part A of the Phase 3 ATTRibute-CM study in late 2021 or early 2022, and from Part B in 2023. If the change in 6MWD is highly statistically significant, we anticipate submitting an NDA for ATTR-CM in 2022. In Part B, the study will continue for a total duration of 30 months, at which point all-cause mortality and cardiovascular hospitalizations will be compared between treatment and control groups. A schematic of the trial is shown below:

 

 

 

Market Opportunity

We believe that the total market for ATTR therapeutic interventions will continue to grow for the foreseeable future as the population of diagnosed patients increases as a result of heightened disease awareness and the adoption of non-invasive diagnostic techniques. As such, if acoramidis is approved, we believe that there could be a significant population of newly diagnosed patients that can be treated with acoramidis who have not previously been treated with a disease-modifying therapy. If approved, we believe that acoramidis could have meaningful commercial potential. Further, we believe that acoramidis, if approved, has the potential to demonstrate benefit as a best-in-class stabilizer for the treatment of ATTR.

9


Competition

If acoramidis is approved as a treatment for ATTR-CM, we expect to face competition primarily from Vyndaqel / Vyndamax (tafamidis meglumine/tafamidis), which is approved in the United States, European Union and Japan as a treatment for ATTR-CM. Additionally, there are a number of RNAi, antisense oligonucleotide and gene editing product candidates that are currently in development as potential treatments for ATTR-CM.

Low-dose Infigratinib: Achondroplasia

Summary

We are developing low-dose infigratinib, an oral FGFR1-3 selective tyrosine kinase inhibitor, or TKI, for the treatment of achondroplasia. While we are also investigating infigratinib as a potential treatment for certain oncology indications, we are utilizing a significantly lower dose in children with achondroplasia than we are for our oncology programs. We are currently enrolling patients in PROPEL, a prospective observational study in children with achondroplasia and PROPEL 2, a Phase 2 dose-escalation and expansion study of infigratinib in children with achondroplasia. We anticipate that we will report initial data from PROPEL 2 in the second half of 2021.

Condition Overview

Achondroplasia is the most frequent cause of disproportionate short stature, and FGFR3 mutations have been shown to be the molecular source of the condition. Achondroplasia has a prevalence of greater than 55,000 in the United States and European Union, and an estimated worldwide incidence of one in 10,000 to 30,000 live births. The condition leads to a disproportionate short stature with anomalies in bone development and potential for foramen magnum stenosis, spinal stenosis, cardiovascular complications and obesity. The average height is approximately 4’4” for a male and 4’1” for a female with achondroplasia. Lifespan and intelligence are most often normal.

Achondroplasia is an autosomal dominant condition caused by a gain-of-function point mutation in the FGFR3 gene. Approximately 97% of cases are due to G380R substitution and 80% of cases are the result of de novo mutations. FGFR3 is expressed in osteoblasts and chondrocytes where it plays a critical role in regulating bone growth through the MAPK pathway, which drives hypertrophic differentiation, and through the STAT1 pathway, which drives chondrocyte proliferation. Apart from growth hormones, which are approved in Japan, we are not aware of any other medicines approved for marketing by the FDA or the European Medicines Agency, or the EMA, for the treatment of achondroplasia.

Our Product Concept

FGFR3 gain-of-function mutations are the driver behind the pathophysiology of achondroplasia. As an FGFR1-3 inhibitor, we believe that infigratinib has the potential to decrease pathologic signaling downstream of FGFR3 and treat achondroplasia at its source. Unlike potentially competitive CNP mimetic approaches, which only inhibit MAPK signaling, our approach also inhibits STAT1 signaling.

Preclinical proof-of-concept has been demonstrated in an achondroplasia mouse model at dose levels as low as 2% of those used in our oncology trials. In our Phase 1 dose escalation clinical trials of infigratinib, we saw acceptable tolerability, including no instances of hyperphosphatemia, at three to six times the expected dose level in our achondroplasia trials. Based on these results, we do not expect significant tolerability issues at the proposed dose level in the clinic.

Preclinical Data

Infigratinib has been studied preclinically in a mouse model of achondroplasia that recapitulates anomalies of the growth plates, vertebrae, and intervertebral discs. Investigators observed that infigratinib rescued ex vivo bone growth of mutant mouse embryo femurs after six days of treatment. Further, 15 days of treatment showed in vivo bone growth, which mimics human achondroplasia in many respects. Effects on both appendicular and axial skeletal parameters were observed in this study. 

10


Below are figures demonstrating the extent of femur growth and intervertebral disc width rescue in wild-type, untreated model, and infigratinib treated (2 mg/kg) model mice:

 

 

In vivo bone growth was further demonstrated at lower doses (0.2 mg/kg and 0.5 mg/kg) by the same laboratory. Together, preclinical studies at all doses have demonstrated meaningful increases in skeletal growth parameters between treated and untreated mutant mice, as follows:

lncrease in length compared to non-treated mouse (%)

 

 

Notably, treatment with infigratinib did not modify the expression of FGFR1 in the hypertrophic zone of the growth plate. The effects seen were mainly due to FGFR3 inhibition, with no other gross side effects being observed in these preclinical studies. Furthermore, survival was improved after 15 days in infigratinib treated mice, regardless of dose, as compared to untreated mice.

Clinical Development Plan

We are currently enrolling patients in PROPEL, a prospective observational study in children with achondroplasia. The study will establish annualized growth velocity, or AGV, for each child for a minimum period of six months. PROPEL is designed to provide baseline measurements for children that we anticipate enrolling in PROPEL 2, an ongoing Phase 2 study of low-dose infigratinib.

11


PROPEL 2 is designed as an open-label, dose-escalation and expansion trial in children with achondroplasia prior to growth plate closure. The primary objective of this study is to assess safety and tolerability in children with achondroplasia. Secondary objectives will include PK analyses, change in growth velocity, and assessment of quality of life. We anticipate that we will report initial data from PROPEL 2 in the second half of 2021.

Key Competitors

Infigratinib is the only oral direct FGFR1-3 inhibitor that has been publicly disclosed in development for the treatment of achondroplasia. There are four other identified companies developing compounds for the treatment of achondroplasia using alternative mechanistic approaches: BioMarin Pharmaceutical Inc. (vosoritide), Ascendis Pharma A/S (TransCon CNP), Pfizer Inc. (recifercept) and Sanofi S.A. (SAR442501).

BBP-631: Congenital Adrenal Hyperplasia

Summary

We are developing BBP-631, a preclinical AAV gene transfer product candidate, for the treatment of CAH, caused by 21OHD. BBP-631 was granted orphan drug designation from both the FDA for the treatment of congenital adrenal hyperplasia 21-hydroxylase deficiency and the EMA for the treatment of congenital adrenal hyperplasia in 2018. We anticipate initiating a Phase 1/2 first-in-human clinical trial of BBP-631 in the second half of 2021, with initial data from this study anticipated in late 2021 or early 2022.

Disease Overview

CAH is a debilitating and life-threatening disease with no available cure, despite newborn screening for the disease being conducted in every U.S. state. The disease is defined by an inability to produce cortisol and aldosterone, and an excess production of testosterone. Lack of cortisol disrupts glucose metabolism and the body’s normal response to stress, leading to potentially fatal adrenal crises, while lack of aldosterone disrupts sodium retention, resulting in low blood pressure, arrhythmia and dehydration. Additionally, excess testosterone causes virilization in females, often leading to ambiguous genitalia and masculinizing features at birth. Hormonal changes during puberty compound the CAH deficiencies. Females often suffer from limited fertility and require intensive treatment before, during and after pregnancy, and up to 40% of adult males will have adrenal rest tumors that can lead to gonadal dysfunction and infertility, occasionally requiring surgery.

Over 90% of CAH cases are caused by 21OHD, a genetic defect in the CYP21A2 gene coding for the enzyme 21OH. Mutations resulting in loss of enzymatic activity of 21OH prevent conversion of progesterone into 11-deoxycorticosterone and 17-hydroxyprogesterone (17OHP) into 11-deoxycortisol, which are the precursors to aldosterone and cortisol, respectively.

CAH patients with 21OHD can be divided into two categories depending on the type of genetic mutation: classic and non-classic. We are primarily focused on treating classic patients, who have the more severe phenotype and that can be categorized into simple virilizing (approximately 25% of patients) and salt-wasting (approximately 75%) by the severity of aldosterone deficiency and level of residual 21OH enzyme activity. Patients with the salt-wasting form of disease have residual enzyme activity of 0% to 1% of normal and patients with the simple virilizing phenotype have 1% to 10% enzyme activity. All patients with the classic form require treatment at birth, as cortisol deficiency can lead to adrenal crisis as early as one to four weeks of life and can quickly lead to death. The salt-wasting form has an incidence of one in 20,000 births, while the simple virilizing form has an incidence of one in 60,000 births. Together, these translate to an estimated 600 classic patients born in the United States and Europe per year. We estimate there are more than 75,000 patients in the United States and Europe in the total addressable patient population.

Current standard of care treatments do not cure patients, but replace missing glucocorticoids, such as cortisol, and mineralocorticoids, such as aldosterone, as well as reduce excessive androgen secretion. Although glucocorticoids are the mainstay of CAH therapy, individuals respond in varying ways, and chronic use of glucocorticoids in children and adults requires careful management because of the well-known side effects of these drugs, such as Cushingoid features, metabolic disease, obesity, hypertension, growth retardation, glucose intolerance, electrolyte disturbance, bone demineralization/increased risk of fracture and delayed puberty. Clinical management of classic CAH is often a very difficult balance between hyperandrogenism and hypercortisolism.

12


Our Product Concept

BBP-631 is a preclinical intravenously administered AAV5 gene transfer product candidate designed for the treatment of CAH due to 21OHD by replacing the 21OH enzyme in the adrenal cortex. Replacement of enzyme function has the potential to normalize flux through the pathway, simultaneously addressing the lack of cortisol and aldosterone, as well as the excess of testosterone and other androgens. Genotype-phenotype correlation studies in CAH suggest that non-classic patients, who are often asymptomatic and do not require treatment, have enzyme activity that is a little as 10% to 20% of normal individuals. We believe that an AAV gene therapy may be able to restore this level of enzymatic activity in CAH patients with both simple virilizing and salt-wasting forms of disease, providing substantial clinical impact and potentially eliminating the need for treatment with exogenous steroids. BBP-631 was granted both FDA and EMA orphan drug designation in 2018 for the treatment of CAH caused by 21OHD.

Preclinical Data

Initial preclinical activity was explored in a Cyp21 knockout mouse model using AAVrh10. An intravenous, or IV, injection of vector genomes was observed to improve multiple disease-related factors over a 15-week duration window, including an increase in body weight, a decrease in urinary progesterone (the main substrate of 21OH), and an increase in renin expression (signaling an increased capacity for salt retention).

A study in non-human primates, or NHP, comparing evaluated AAV serotypes 1, 5 and 6 identified AAV5 as the optimum serotype. We observed significant transfection in the adrenals where 21OH is synthesized. Additionally, AAV5 has relatively low seroprevalence in the human population, which may limit potential immunogenicity issues.

We have completed two sets of NHP studies designed to evaluate durability of expression, dosing/transgene expression relationships and preliminary safety. In the first set of experiments, which evaluated a lower dose of 3x10 12 vector genomes per kilogram, we observed sustained increases in Cyp21 mRNA levels up to six months out. We did not observe rapid decreases in vector genome counts and mRNA levels due to adrenal cell turnover between 1.5 and six months, providing preliminary support for sustained transgene expression.

In a second set of experiments, a total of 20 NHPs were treated with BBP-631 at one of three IV doses. Vector copy number and transgene mRNA expression in the adrenal glands were analyzed at four and 12 weeks post-dosing in the low- and medium-dose arms, and at 12 and 24 weeks post-dosing in the high-dose arm. No dose-related AEs were observed at any of the doses tested at any time point.

Overall, treatment with BBP-631 resulted in high vector copy number, or VCN, and mRNA expression in the adrenal gland, suggesting strong tropism and uptake of BBP-631 for the adrenal gland. In the high-dose arm, VCNs were maintained between 12 and 24 weeks. Furthermore, mRNA levels increased between 4 and 12 weeks for the medium dose arm and were consistent between 12 and 24 weeks for the high dose arm. Researchers also saw dose-dependent increases in both VCNs and mRNA levels across the three doses tested.

Key Competitors

There are two alternative therapeutic classes being investigated for treatment of CAH. The first are corticotropin-releasing factor type 1, or CRF1, receptor antagonists. CRF1 receptor antagonists regulate the release of adrenocorticotropic hormone, or ACTH, from the pituitary gland, which stimulates androgen and cortisol synthesis in the adrenal gland. In healthy individuals, endogenous cortisol provides negative feedback to the release of ACTH, which keeps androgen synthesis well regulated. Because this negative feedback is severely impaired in CAH patients, supraphysiologic doses of exogenous steroids are required to normalize androgen synthesis in these patients. While CRF1 receptor antagonists may regulate androgen synthesis, they do not address the lack of cortisol or aldosterone production in these patients. Therefore, steroid supplementation is still required with CRF1 receptor antagonists. Two CRF receptor antagonists, Crinecerfont (under development by Neurocrine Biosciences, Inc.) and Tildacerfont (under development by Spruce Biosciences, Inc.), are currently in Phase 3 and Phase 2b clinical trials, respectively.

13


The second alternative therapeutic class is ACTH receptor antagonists. Inhibition of this pathway, which is downstream of the CRF1 pathway, also results in inhibition of androgen and cortisol synthesis in the adrenal gland. However, like CRF1 receptor antagonists, ACTH inhibitors do not address the lack of cortisol or aldosterone production in these patients. CRN04904, an oral ACTH antagonist, is currently in Phase 1 clinical development by Crinetics Pharmaceuticals, Inc.

While these alternative therapeutic mechanisms attempt to address meaningful aspects of the disease by potentially reducing the need for exogenous steroids, neither is able to address the disease at its source by targeting the complete set of features that define the disease. In particular, these mechanisms cannot obviate the need to administer steroids because they do not address the body’s inability to synthesize cortisol and aldosterone. In contrast, we believe enzymatic replacement by gene therapy has the potential to simultaneously address all facets of the disease by restoring proper flux through the hormonal pathways, reducing androgen production by providing alternative pathways for the precursor molecules to be converted into cortisol or aldosterone.

Encaleret: Autosomal Dominant Hypocalcemia Type 1 and Hypoparathyroidism

Encaleret is an oral small molecule antagonist of the calcium sensing receptor, or CaSR, that we are developing for the treatment of Autosomal Dominant Hypocalcemia Type 1, or ADH1. We are currently studying encaleret in an ongoing Phase 2b clinical trial as a potential treatment for patients with ADH1, and anticipate reporting early results from this study in the first half of 2021. Encaleret has been granted orphan drug designation as a treatment for autosomal dominant hypocalcemia, including ADH Type 1 and ADH Type 2.

Hypoparathyroidism, or HP, is a disease in which the parathyroid gland secretes no or abnormally low levels of parathyroid hormone, or PTH, which results in hypocalcemia. Common presenting symptoms of hypoparathyroidism, related to hypocalcemia, including muscle cramps, carpopedal spasms, tingling and seizures. ADH1 is a rare, genetic form of HP caused by gain-of-function mutations of the CaSR, and which is characterized by increased sensitivity of the CaSR to calcium level. Symptoms due to hypocalcemia can be more severe than in other forms of HP, and include severe muscle cramping, seizures and kidney damage resulting from hypercalcuria. ADH1 has a prevalence of approximately 12,000 variant carriers in the United States. Chronic hypoparathyroidism has a larger estimated prevalence of approximately 200,000 in the United States and the European Union. No FDA or EMA approved therapies for ADH1 currently exist, although hypocalcemia is typically managed with calcium and activated vitamin D supplementation. Natpara (parathyroid hormone) was approved by the FDA in 2015 as an adjunct to calcium and vitamin D to control hypocalcemia in patients with hypoparathyroidism, and has a labeled limitation of its use in patients with ADH1.

Our Product Concept

Encaleret is an investigational small molecule antagonist of the CaSR. It has been studied in more than 1,200 human subjects and was observed to increase serum calcium in a dose-dependent manner. The rationale for developing encaleret as a potential treatment for patients with ADH1 is based on both non-clinical and clinical evidence. Antagonists of the CaSR have been shown, in both in vitro and in vivo models, to shift the aberrant CaSR “set-point” back towards a normal IC50 for calcium, resulting in increased PTH secretion, elevation of blood calcium concentrations, and reduction of urinary calcium excretion in cellular and animal models of ADH1. By selectively antagonizing the CaSR, encaleret may restore normal CaSR function in individuals with ADH1 and may address symptoms associated with hypocalcemia and hypercalciuria.

Clinical Development Plan

We have initiated a single-center Phase 2b study investigating encaleret in ADH1 at the National Institutes of Health. We anticipate reporting early results from this study in the first half of 2021.

Competitors

Encaleret is the only molecule that has been publicly disclosed in development for the treatment of ADH1. There are other identified companies developing compounds for the treatment of hypoparathyroidism using

14


recombinant parathyroid hormone analogs or PTH receptor agonists: Takeda Pharmaceutical Company (Natpara), Ascendis Pharma A/S (TransCon PTH), Amolyt Pharma (AZP-3601), Chugai Pharmaceutical Company (PCO371), and Extend Biosciences Inc. (EXT607).

OTHER DEVELOPMENT PROGRAMS

MENDELIAN PORTFOLIO

Fosdenopterin: MoCD Type A

We are developing fosdenopterin, an IV formulation of synthetic cyclic pyranopterin monophosphate, or cPMP, for the treatment of molybdenum cofactor deficiency, or MoCD, Type A. Fosdenopterin received breakthrough therapy designation from the FDA in 2013 for MoCD, orphan drug designation from the FDA in 2009 and EMA in 2010 for the treatment of MoCD Type A, and rare pediatric disease designation for the treatment of MoCD Type A in June 2017. We filed an NDA for fosdenopterin with the FDA in 2020, which has been granted Priority Review designation.

MoCD Type A is an ultra-rare autosomal recessive inborn error of metabolism caused by disruption in molybdenum cofactor, or MoCo, biosynthesis which results in deficiencies in multiple enzyme activities, including sulfite oxidase, or SOX, and leads to uncontrolled sulfite toxicity in the brain. The disease typically presents very early in life, with median presentation at first day of life. The disease is characterized by severe and rapidly progressive acute sulfite-related neurological damage and associated heterogeneous neurological sequelae including seizures, feeding difficulties and in most cases death, with the median survival estimated to be approximately three years. Incidence is estimated to be one in 100,000 to 200,000 live births worldwide, with MoCD Type A accounting for approximately two-thirds of all cases. There are no available treatments approved for any form of MoCD. Supportive care and anti-convulsant therapy may be used to manage symptoms.

BBP-671: PKAN and Organic Acidemias

BBP-671 is an oral, small molecule, allosteric activator of pantothenate kinases that we are developing for the treatment of Pantothenate Kinase Associated Neurodegeneration, or PKAN, and Organic Acidemias, or OAs. BBP-671 is currently in preclinical development. BBP-671 has received orphan drug designation as a treatment of proprionic acidemia, or PA, and as a treatment of PKAN. BBP-671 was also designated as a drug for a rare pediatric disease for treatment of both PKAN and PA.

PKAN is a rare genetic disorder with progressive neurodegeneration. Early onset patients typically demonstrate motor deficits with possible visual problems from retinal degeneration within six years of age. Later onset disease is heterogeneous, with psychiatric symptoms and progressive parkinsonism developing in late childhood to adulthood. The prevalence of PKAN is approximately one in 1,000,000, with between 800 to 850 patients in the United States and the European Union. There are currently no approved treatments for PKAN.

OAs are caused by mutations in enzymes that disrupt amino acid metabolism leading to acute decompensations requiring hospitalization, as well as long-term complications involving multiple organ systems, such as the heart, pancreas, kidney, liver and brain. The incidence of OAs is approximately five in 100,000 births. The  standard of care includes dietary restriction and supplementation, but unmet need remains high due to metabolic decompensations and long-term complications.

BBP-711: Primary Hyperoxaluria and Frequent Stone Formers

BBP-711 is an oral, small molecule inhibitor of glycolate oxidase, or GO, that we are developing for the treatment of primary hyperoxaluria and patients who experience frequent kidney stone formation. BBP-711 is currently in preclinical development. BBP-711 has received orphan drug designation from the FDA as a treatment of primary hyperoxaluria type 1, or PH1. BBP-711 has also been designated as a drug for a rare pediatric disease for treatment of PH1.

PH1 is a rare, autosomal-recessive inborn error of metabolism driven by a defect in the AGXT gene, which codes for the enzyme alanine-glyoxylate aminotransferase, or AGXT.  Deficiencies in the AGXT enzyme translate into the incapacity of PH1 patients to detoxify glyoxylate into glycine. As a result, glyoxylate is oxidized into oxalate, which cannot be metabolized by humans. Elevated oxalate levels form calcium oxalate crystals, and

15


subsequently kidney stones, which damage the kidneys, culminating in renal dysfunction. Prevalence for PH1 is estimated to be 5,000 patients in the United States and the European Union. Due to heterogeneous symptom presentation and similarity with other diseases, we believe that the disease is underdiagnosed. Prevalence for frequent stone formers is estimated to be 1.5 million in the United States and the European Union. Standard of care involves symptomatic management through supplementation with vitamin B6, increased fluid intake, and citrate, to intensive dialysis and lithotripsy. Ultimately, the only curative treatment is a combined liver and kidney transplant.

BBP-418: Limb Girdle Muscular Dystrophy Type 2i

BBP-418 is an orally administered ribitol replacement therapy we are developing for the treatment of Limb Girdle Muscular Dystrophy type 2i, or LGDM2i. We are currently studying BBP-418 in an ongoing Phase 1 clinical trial. Subject to successful completion of the Phase 1 clinical trial, we anticipate initiating a Phase 2 clinical trial in 2021 and generating top-line data from this clinical trial in 2022.

LGMD2i is an inherited rare progressive genetic disorder characterized by lower-limb weakness and loss of ambulation, in addition to potential pulmonary and cardiac dysfunction. LGMD2i has an estimated prevalence of around 4.5 per 1,000,000. There is no disease-modifying treatment available. Standard of care for fukutin-related protein gene dystroglycanopathies is supportive care to alleviate end organ dysfunction.

BBP-472: PI3KB Inhibitor for Autism-Spectrum Disorder Characterized by Loss of PTEN Protein

BBP-472 is a series of small molecule PI3KB inhibitors being designed to balance kinase signaling in the brain for the treatment of children with autism-spectrum disorders characterized by loss of the PTEN protein. BBP-472 is currently in preclinical development.

GENETIC DERMATOLOGY

BBP-009/Patidegib (PellePharm): Gorlin Syndrome and High Frequency Basal Cell Carcinoma

BBP-009 is a topical gel formulation of patidegib, a hedgehog inhibitor, that we are developing for the treatment of Gorlin Syndrome and High-Frequency Basal Cell Carcinoma, or HF-BCC. We are currently conducting a Phase 3 clinical trial of BBP-009 in Gorlin Syndrome and a Phase 2b clinical trial of BBP-009 in HF-BCC. We have received breakthrough therapy designation from the FDA, as well as orphan drug designation from both the FDA and EMA, for BBP-009 for the treatment of Gorlin Syndrome.

In November 2018, through our investment in PellePharm, Inc., or PellePharm, we entered into a partnership with LEO Pharma A/S, or LEO, pursuant to which LEO has acquired a minority stake in PellePharm and has agreed to provide additional non-dilutive capital to fund the development of topical patidegib, including our planned Phase 3 clinical trial. LEO also acquired an option to purchase all shares in PellePharm at a later date. See “—Our Material Agreements—BBP-009 (Patidegib): Option Agreement with LEO Pharma A/S.”

BBP-589/PTR-01: Recessive Dystrophic Epidermolysis Bullosa

We are developing BBP-589, an IV-administered recombinant collagen type VII, or rC7, protein replacement therapy, for the treatment of recessive dystrophic epidermolysis bullosa, or RDEB. BBP-589 received orphan drug designation from the FDA and EMA in 2014 for the treatment of dystrophic epidermolysis bullosa, or DEB, and we received fast track designation from the FDA in 2019 for the treatment of DEB.

We have completed enrollment in both a Phase 1/2 clinical trial and a Phase 2 clinical trial in patients with RDEB. We anticipate that we will provide data from the ongoing Phase 2 study in late 2021 or early 2022.

BBP-681: Venous and Lymphatic Malformations

BBP-681 is a transdermal PI3K inhibitor that we are developing for the treatment of cutaneous venous and lymphatic malformations. We are currently studying BBP-681 in an ongoing Phase 1/2 clinical trial and anticipate providing initial data from this study in 2022.

16


Venous malformations, or VMs, are large, disorganized veins that can cause significant morbidity due to functional impairment, pain, bleeding and disfigurement. Lymphatic malformations, or LMs, involve the lymphatic vessels and cause functional impairment and pain similar to VM, lymphatic leakage and disfigurement. The prevalence of VMs and LMs is greater than 75,000 and 42,000, respectively, in the United States and the European Union in the skin.  Standard of care is generally non-disease-modifying and invasive and ranges from compression bandages and aspirin, to laser ablation, surgical resection and sclerotherapy.  

BBP-561: Netherton Syndrome

BBP-561 is a topical KLK5/7 inhibitor that we are developing for the treatment of Netherton Syndrome. BBP-561 is currently in preclinical development.

Netherton Syndrome is a devastating genetic disease characterized by skin breakdown complicated by risk of sepsis, severe malnutrition, and dehydration in affected neonates. It can additionally lead to chronic problems including allergy, infection and inflammation. The prevalence is approximately 4,000 to 17,000 patients in the United States and the European Union. No disease-modifying therapy exists. Palliative and preventative treatments are used to manage symptoms.

TARGETED ONCOLOGY

Infigratinib: FGFR-Driven Cancers

We are developing infigratinib, an oral FGFR1-3 selective TKI for the treatment of FGFR-driven cancers. Specifically, we are developing infigratinib in three oncologic indications: (i) cholangiocarcinoma, or CCA, or bile duct cancer, with FGFR2 fusions or translocations, (ii) urothelial carcinoma, or UC, with FGFR genomic alterations, and (iii) other cancers with FGFR fusions or translocations.

We filed an NDA with the FDA in late 2020 for infigratinib as a second-line or later therapy in patients with advanced and/or metastatic CCA with FGFR2 fusions or translocations. The FDA has granted our NDA for infigratinib in cholangiocarcinoma Priority Review designation and it is being reviewed under the Real-Time Oncology Review, or RTOR, pilot program, which is an initiative of the FDA’s Oncology Center of Excellence designed to expedite the delivery of safe and effective cancer treatments to patients. We are studying infigratinib as a potential treatment for FGFR-driven cancers in multiple ongoing clinical trials:

 

a Phase 3 randomized, open-label clinical trial of infigratinib as a first-line therapy for CCA compared to gemcitabine and cisplatin in advanced and/or metastatic CCA with FGFR2 fusions or translocations;

 

a Phase 2 single-arm clinical trial in patients with advanced or metastatic cholangiocarcinoma with FGFR alterations;

 

a Phase 3 randomized, double-blind, placebo-controlled clinical trial in cisplatin-ineligible adjuvant UC with FGFR3 genomic alterations; and

 

a Phase 2 single-arm clinical trial in patients with advanced or metastatic solid tumors harboring FGFR1-3 gene fusions or genetic alterations.

We received Fast Track Designation in adults with first-line advanced or metastatic cholangiocarcinoma and the FDA and EMA have granted orphan drug designation for infigratinib as a treatment of cholangiocarcinoma.

BBP-398: Targeting Multiple Oncology Indications

BBP-398 is a small molecule inhibitor of SHP2 that we are developing as a potential treatment of cancers driven by hyperactive receptor tyrosine kinase, or RTK, or MAPK signaling. We are currently enrolling patients in a Phase 1 dose escalation and expansion clinical trial in patients with RAS and RTK mutations.

17


SHP2 is a phosphatase that acts downstream of receptor tyrosine kinases in the MAPK signaling pathway. SHP2 is critical in signaling in these pathways. Increased MAPK signaling is a hallmark of a number of cancer types, including cancers driven by RTK genetic alterations, cancers with RTK fusion mutations, and cancers with constitutively active MAPK signaling. Additionally, SHP2 is implicated as a downstream mediator of PD-1 signaling, a key target of immuno-oncology treatment.

BBP-954: Multiple Oncology Indications

BBP-954 is a preclinical discovery program for irreversible inhibitors of glutathione peroxidase 4, or GPX4, for the treatment of solid and hematological cancers.

Ferroptosis is a form of oxidative programmed cell death that cancer cells must avoid in order to survive and form tumors. GPX4 is an enzyme that protects cancer cells from ferroptosis by neutralizing toxic lipid free radicals. By inhibiting GPX4, we aim to trigger ferroptosis in cancer cells. Preclinical data generated by us and third parties suggest many of the most common cancers are sensitive to GPX4 inhibition, both in monotherapy and combination with standard anti-cancer agents such as kinase inhibitors and chemotherapy. We believe that GPX4 may be potentially applicable to a number of common solid and hematologic cancers, including non-small cell lung cancer, breast cancer, melanoma, pancreatic adenocarcinoma, renal cell carcinoma and Non-Hodgkin’s lymphoma, among others.

BBP-454: KRAS-Driven Cancers

BBP-454 is a preclinical development program focused on approaches to inhibit KRAS through novel selective mechanisms, for the treatment of KRAS-driven cancers.

KRAS is a member of the RAS family of oncogenes, which also includes HRAS and NRAS, and together comprises some of the most well-known monogenic drivers of cancer. Mutations in NRAS are frequently found in leukemia and melanoma, while HRAS is frequently mutated in bladder, thyroid, and head and neck squamous cell carcinoma. KRAS mutations are a frequent driver of a number of the largest cancer indications with high unmet medical need, including 30% of non-small cell lung cancers, 98% of pancreatic adenocarcinomas and 45% of colorectal adenocarcinomas. The most common KRAS mutations involve a change from glycine at position 12 in the protein to aspartic acid (G12D, 36% of all KRAS mutations), valine (G12V, 24%), and cysteine (G12C, 15%), but also include mutations at glycine 13 and glutamine 61. In aggregate, over 500,000 patients in the United States and Europe are diagnosed with KRAS-driven cancers, annually.

KRAS-mutant cancers are driven by active, GTP-bound KRAS located at the cell membrane. We are developing multiple strategies that target KRAS through novel, mutation-agnostic mechanisms. We anticipate nominating our first development candidate from this discovery program in 2022.

GENE THERAPY

BBP-812: Canavan Disease

BBP-812 is an AAV gene therapy product candidate that we are developing for the treatment of Canavan Disease that is designed to deliver the ASPA gene, which is defective in patients with Canavan disease. BBP-812 is currently in preclinical development.  

Canavan Disease is a fatal, progressive neurodegenerative disorder that begins in infancy. The disease is a leukodystrophy, caused by degradation of white matter in the brain. Patients typically miss developmental milestones, have a rapidly increasing head circumference, progressive lack of motor control, and often do not live past their mid-teens. The incidence of Canavan Disease is approximately one in 100,000 births worldwide.  No treatments are approved for Canavan Disease; care is focused on symptom management.

18


BBP-815: TMC1-related Hearing Loss

BBP-815 is an AAV gene therapy product candidate that we are developing for the treatment for nonsyndromic hearing loss caused by recessive mutations in the TMC1 gene. Mutations in the TMC1 gene prevent sound from eliciting the appropriate electrical response in the hair cells, resulting in moderate to severe hearing loss, often present early in life. BBP-815 is currently in preclinical development.

Additional Program-Related Information

Manufacturing

We do not own or operate, and currently have no plans to establish, any manufacturing facilities. We currently depend on third-party contract manufacturing organizations, or CMOs, for all of our requirements of raw materials, drug substance and drug product for our preclinical research and our ongoing clinical trials of our product candidates. Aside from a manufacturing agreement that we entered into in December 2019 through our subsidiary, BridgeBio Gene Therapy, LLC, we have not entered into long-term agreements with our current CMOs. We intend to continue to rely on CMOs for later-stage development and commercialization of our product candidates, including any additional product candidates that we may identify. Although we rely on CMOs, we have personnel and third-party consultants with extensive manufacturing experience to oversee the relationships with our contract manufacturers. Several of our development candidates have or are in the near term expected to have redundant and overlapping drug substance and drug product supply chains.

Sales and Marketing

We intend to begin building a commercial infrastructure in the United States and selected other territories to support the commercialization of each of our product candidates when we believe a regulatory approval in a particular territory is likely. Because most of our target indications are rare diseases with a concentrated prescribing audience and a small number of key opinion leaders who influence the treatments prescribed for the relevant patient population, we currently believe that we can effectively address each market using our own targeted, specialty sales and marketing organization supported by internal sales personnel, an internal marketing group and distribution support.

We intend to evaluate our commercialization strategy as we advance each product candidate through clinical development. In any core markets outside of the United States that we may identify, where appropriate, we may utilize strategic partners, distributors or contract sales forces to expand the commercial availability of our product candidates. We currently do not expect that we will require large pharmaceutical partners for the commercialization of any of our product candidates, although we may consider partnering in certain territories or indications or for other strategic purposes.

Intellectual Property

Overview

We strive to protect the proprietary technology that we believe is important to our business through a variety of methods, including seeking and maintaining patents and patent applications intended to cover our product candidates and compositions, their methods of use and processes for their manufacture, our platform technologies and any other aspects of inventions that are commercially important to the development of our business. We seek to obtain domestic and international patent protection and, in addition to filing and prosecuting patent applications in the United States, we may file counterpart patent applications in additional countries where we believe such foreign filing is likely to be beneficial, including Australia, Canada, Europe, China, Japan, and Mexico. We have entered into various license agreements to obtain the rights to use certain patents for the development and commercialization of our product candidates. See “—Our Material Agreements.” We also rely on trade secrets to protect aspects of our business that are not amenable to, or that we do not consider appropriate for, patent protection.

Our success will depend on our ability to obtain and maintain patent and other proprietary rights protecting our commercially important technology, inventions and know-how related to our business, defend and enforce our current and future issued patents, if any, preserve the confidentiality of our trade secrets and operate without

19


infringing the valid and enforceable patents and proprietary rights of third parties. We continually assess and refine our intellectual property strategy in order to best fortify our position, and file additional patent applications when our intellectual property strategy warrants such filings. We also rely on know-how, continuing technological innovation and potential in-licensing opportunities to develop and maintain our intellectual property portfolio. We seek to obtain domestic and international patent protection, and endeavor to promptly file patent applications for new commercially valuable inventions.

The patent positions of biopharmaceutical companies like us are generally uncertain and involve complex legal, scientific and factual questions. In addition, the coverage claimed in a patent application can be significantly reduced before the patent is issued, and patent scope can be reinterpreted by the courts after issuance. Moreover, many jurisdictions permit third parties to challenge issued patents in administrative proceedings, which may result in further narrowing or even cancellation of patent claims. We cannot predict whether the patent applications we are currently pursuing will issue as patents in any particular jurisdiction or whether the claims of any patents, if issued, will provide sufficient protection from competitors.

Because patent applications in the United States and certain other jurisdictions are maintained in secrecy for 18 months or potentially even longer, and since publication of discoveries in the scientific or patent literature often lags behind actual discoveries, we cannot be certain of the priority of inventions covered by pending patent applications. Moreover, we may have to participate in interference proceedings or derivation proceedings declared by the United States Patent and Trademark Office, or USPTO, to determine priority of invention.

As of February 15, 2021, our intellectual property portfolio is composed of 65 issued patents and 135 patent applications that we license from academic and research institutions and other third parties, and 25 issued patents and 220 pending patent applications that we own, including through our subsidiaries. These patents and patent applications generally provide us with the rights to develop our product candidates in the United States and worldwide. Our intellectual property portfolios for each of the programs that we consider to be our core value drivers are further described below.

QED Therapeutics, Inc.

 

For our subsidiary, QED Therapeutics, Inc., we license rights from Novartis to two issued U.S. patents, and related pending and issued foreign patents and patent applications in Australia, Canada, China, Europe, Japan and Mexico, as well as in other countries in Asia and in South America, that are directed to compositions of matter of BBP-831. The issued U.S. patents are expected to expire between 2026 and 2029, which takes into account patent term adjustments granted by the USPTO. The foreign patents and patent applications, if issued, are expected to expire between 2025 and 2030.

 

We also license rights from Novartis to one issued U.S. patent, one pending U.S. patent application, and related pending and issued foreign patents and patent applications in Australia, Canada, China, Europe, Japan and Mexico, as well as in other countries in Asia and in South America, that are directed to pharmaceutical formulations containing BBP-831. The issued patents and patent applications, if issued, are expected to expire in 2034.

 

We also license rights from Inserm Transfert ESA and Assistance Publique-Hôpitaux de Paris to one issued U.S. patent and one pending U.S. patent application, and one granted patent in Europe, that are directed to methods of treating achondroplasia using BBP-831. The issued U.S. patent, granted patent in Europe, and the pending patent application, if issued, are expected to expire in 2032.

 

In addition, QED Therapeutics, Inc. owns one pending U.S. provisional patent application and two pending PCT patent applications that are directed to methods of treating various cancers using BBP-831. If any patents issue from these patent applications, such patents would be expected to expire in 2040 or 2041.

20


Eidos Therapeutics, Inc.

 

For our subsidiary Eidos Therapeutics, Inc., we license rights from the Board of Trustees of the Leland Stanford Junior University, or Stanford, to nine issued U.S. patents with claims directed to composition of matter and methods of use relating to acoramidis. These patents are expected to expire in 2031 or 2033. We also license rights from Stanford to two pending U.S. patent applications, one issued European patent, one pending European patent application, and one issued Japanese patent with claims directed to composition of matter and methods of use relating to acoramidis. These patents and patent applications, if issued, are expected to expire in 2031 or 2033.

 

In addition, we own one issued U.S. patent, three pending U.S. patent applications, and 50 related foreign patent applications pending in various jurisdictions, including Australia, Canada, Europe, China, Japan, and Mexico with claims directed to salt and solid forms, methods of manufacturing, dosing methods, and formulations relating to acoramidis. The issued U.S. patent is expected to expire in 2038. The pending U.S. and foreign patent applications, if issued, are expected to expire in 2038 or 2039.

Adrenas Therapeutics, Inc.

 

For our subsidiary Adrenas Therapeutics, Inc., we own one pending U.S. patent application and six related foreign patent applications pending in various jurisdictions including Canada, China, Europe, Japan, and Korea with claims directed to recombinant AAV vectors relating to BBP-631. These patent applications, if issued, are expected to expire in 2039.

Phoenix Tissue Repair, Inc.

 

For our subsidiary Phoenix Tissue Repair, Inc., we license rights from the University of Southern California, or USC, to one issued U.S. patent with claims directed to polypeptides comprising functional fragments of collagen 7, and three pending U.S. patent applications with claims directed to methods of use, including treating epidermolysis bullosa with collagen 7. The issued U.S. patent is expected to expire in 2035 and the pending U.S. patent applications, if issued, are expected to expire between 2027 and 2035. We also license rights from USC to five related foreign patents issued in various jurisdictions including Australia, Europe and Japan, and three related foreign patent applications pending in various jurisdictions including Australia. The foreign patents and patent applications, if issued, are expected to expire between 2027 and 2035.

 

We also own six issued U.S. patents, four pending U.S. patent applications, one pending PCT patent application, seven foreign patents, and over 20 foreign patent applications pending in various jurisdictions with claims relating to collagen 7 materials and methods relating to the same. These include issued U.S. patents in the United Kingdom, France and Germany with claims directed to collagen 7 modification for enhancing the degradability of collagen that are expected to expire in 2022. This portfolio also includes pending and/or issued claims relating to collagen 7 detection assays, methods of making and purifying collagen 7, methods of treating subjects having age-related disorders with collagen 7, collagen 7 compositions, and methods of treating epidermolysis bullosa with collagen 7.

 

This portfolio also includes pending and/or issued claims relating to collagen 7 detection assays, methods of making and purifying collagen 7, methods of treating subjects having age-related disorders with collagen 7, collagen 7 compositions, and methods of treating epidermolysis bullosa with collagen 7. These patents and patent applications, if issued, are expected to expire between 2025-2040.

TheRas, Inc.

 

For our subsidiary TheRas, Inc., we license rights from The Regents of the University of California, or the University of California, and Leidos Biomedical Research, Inc., or Leidos, to an issued U.S. patent and two pending U.S. patent applications with claims directed to modulators of K-RAS, which include claims to the modulators as composition of matter and their use in therapy, including the treatment of cancer, and over twenty related foreign patent applications pending in various jurisdictions, including Australia, Canada, China, Europe, Japan, and Mexico. The U.S. patent and the U.S. and foreign patent applications, if issued, are expected to expire in 2036 and 2038. We also co-own with, and license rights from, the University of California and Leidos, three pending Taiwanese patent

21


applications. If issued, any patent applications claiming the benefit of these applications are expected to expire in 2039.

 

In addition, TheRas co-owns with Leidos and Lawrence Livermore National Security, LLC, or Livermore, one pending U.S. patent application with claims directed to modulators of K-RAS, which include claims to the modulators as composition of matter and their use in therapy, including the treatment of cancer. Any patents issuing from this application are expected to expire in 2042.

Our Material Agreements

Acoramidis

License Agreement with Alexion

In September 2019, through our subsidiary Eidos Therapeutics, Inc., or Eidos, we entered into a license agreement, or the “Alexion License Agreement, with Alexion Pharma International Operations Unlimited Company, a subsidiary of Alexion Pharmaceuticals, Inc. , or together, Alexion, to develop and commercialize acoramidis in Japan. Additionally, in September 2019, Eidos entered into a stock purchase agreement with Alexion, pursuant to which Eidos sold to Alexion 556,173 shares of its common stock for aggregate cash proceeds of $25.0 million. Under the terms of the Alexion License Agreement, Eidos granted Alexion an exclusive license to certain of our intellectual property rights to develop, manufacture and commercialize acoramidis in Japan. In consideration for the license grant, Eidos received an upfront payment of $25.0 million, with the potential for an additional one-time payment of $30.0 million subject to the achievement of a regulatory milestone. In addition, Eidos is entitled to receive royalties in the low double-digits on net sales by Alexion of acoramidis in Japan. The royalty rate is subject to reduction if Alexion is required to obtain intellectual property rights from third parties to develop, manufacture or commercialize acoramidis in Japan, or upon the introduction of generic competition into the market.

License Agreement with the Board of Trustees of the Leland Stanford Junior University

In April 2016, through Eidos, we entered into an exclusive license agreement with Stanford for rights relating to novel transthyretin aggregation inhibitors. Under our agreement, Stanford has granted us an exclusive worldwide license to make, use and sell products that are covered by the licensed patent rights. This license grant expires when the last licensed patent expires. The patent rights exclusively licensed to us under the license are described in more detail above under the heading “—Intellectual property— Eidos Therapeutics, Inc.”

Stanford retains the right, on behalf of itself and all other non-profit academic research institutions, to practice under the patent rights for any non-profit purpose, including sponsored research and collaborations. We may grant sublicenses to third parties so long as we are actively pursuing the development or commercialization of products covered by the patent rights. We may also be required to sublicense our rights under the agreement at Stanford’s request under certain conditions, including if we are unwilling or unable to serve a potential market or territory and there is a third party willing to be a sublicensee in such market or territory.

We are obligated to pay to Stanford a yearly license maintenance fee during the term of the agreement, but we may offset the maintenance fee against earned royalty payments due on net sales occurring in that year. Stanford is entitled to receive a royalty as a percentage of net sales of licensed products, in the low single digits. We have agreed to pay Stanford a percentage of non-royalty revenue we receive from our sublicensees, with the amount owed decreasing annually for three years based on when we enter into the applicable sublicense agreement. In addition, we are obligated to pay Stanford up to approximately $1.0 million upon the achievement of specific intellectual property, clinical and regulatory milestone events. In the event of a change of control transaction with respect to Eidos, we are obligated to pay Stanford a change of control fee of $250,000 in connection with the assignment of the license agreement to the acquirer of Eidos.

Under the license agreement with Stanford, we are obligated to use commercially reasonable efforts to develop, manufacture, and commercialize at least one licensed product; to develop markets for such licensed products; and to meet certain development milestones as agreed upon between us and Stanford.

22


Subject to the expiration of the license grant described above, the agreement does not have a specified term. We may terminate the agreement by providing prior written notice to Stanford, and Stanford has the right to terminate the agreement if we fail to achieve certain milestones or make payments under the agreement, or are not actively pursuing development of a licensed product, or if we otherwise materially breach the agreement and fail to cure such breach within a specified grace period.

BBP-831: License Agreement with Novartis International Pharmaceutical Ltd.

In January 2018, through our subsidiary QED Therapeutics, Inc., or QED, we entered into a license agreement with Novartis International Pharmaceutical Ltd., or Novartis, for certain intellectual property rights, including patents and know-how, related to BBP-831 for the treatment of patients with FGFR-driven diseases, including CCA, UC and achondroplasia. We refer to this agreement as the Novartis License.

Pursuant to the Novartis License, we obtained a license to research, develop, make, have made, use, import, offer for sale, sell, have sold and otherwise commercialize BBP-831, as well as therapeutic products incorporating BBP-831 that would, but for the license grant, infringe Novartis’ license patent rights, or that were developed using or that incorporate or embody Novartis’ licensed know-how, in all fields of use worldwide. The license grant to us includes the right to sublicense through multiple tiers. We also have certain rights to intellectual property licensed to Novartis’ affiliate under a materials transfer agreement with a third party.

The Novartis License is subject to Novartis’ existing obligations to supply a third party with BBP-831 to support the third party’s clinical trials, and we have an ongoing obligation to inform Novartis of our or our sublicensees’ intent to seek regulatory approval for and commercialize BBP-831 for various indications, with potential reversionary rights to Novartis in the event of a subsequent decision not to seek regulatory approval and commercialization, or a determination by Novartis that we have failed to sufficiently pursue regulatory approval and commercialization, for Novartis to grant such third party limited rights to develop and commercialize BBP-831.

Under the terms of the Novartis License, we made a one-time payment of $15.0 million to Novartis and agreed to issue shares of Series A preferred stock of QED valued at approximately $1.7 million in the aggregate to Novartis. In addition, we are obligated to make contingent milestone payments totaling $60.0 million upon achievement of certain regulatory milestones. We are also obligated to make contingent milestone payments totaling $35.0 million upon achievement of certain sales milestones for therapeutic products incorporating BBP-831. QED also agreed to pay Novartis tiered low double-digit royalties on net sales of therapeutic products incorporating BBP-831.

Under the Novartis License, we are required to use commercially reasonable efforts to develop BBP-831, and to obtain regulatory approval for and commercialize BBP-831 in the United States and the European Union.

We may terminate the Novartis License in its entirety or on a product-by-product or country-by-country basis at any time with 60 days’ prior written notice to Novartis. Novartis may terminate if QED ceases to function as a going concern, is the subject of certain bankruptcy or similar proceedings, or otherwise winds down or discontinues its business. Either party may terminate for material breach that is not cured by the other party within a specified time period of receiving notice of such material breach. Otherwise, the Novartis License terminates on a product-by-product and country-by-country basis on the latest of the expiration of licensed patent rights, the expiration of regulatory exclusivity, or the tenth anniversary of the first commercial sale in such country.

BBP-870: Asset Purchase Agreement with Alexion Pharma Holding Unlimited Company

In June 2018, through our subsidiary Origin Biosciences, Inc., we entered into an asset purchase agreement with Alexion Pharma Holding Unlimited Company, or Alexion Pharma, pursuant to which we acquired Alexion’s right, title and interest in certain assets relating to fosdenopterin, including patents and other intellectual property rights.

In the event that a Priority Review Voucher, or PRV, is granted to us by the FDA, we have agreed to pay Alexion Pharma a percentage in the mid-teens of any proceeds received by us from our sale of the PRV to a third

23


party. If we do not sell the PRV to a third party within 180 days after our receipt of the PRV, we are obligated to pay Alexion Pharma $18.8 million, which amount is creditable against any amounts otherwise due to Alexion Pharma in accordance with the preceding sentence upon any future sale by us of the PRV. We are obligated to make contingent milestone payments totaling $3.0 million upon achievement of certain development milestones and $17.0 million upon achievement of certain sales milestones for products containing the fosdenopterin molecule. We also agreed to pay Alexion Pharma tiered royalties ranging from the low-to mid-teens on net sales of products containing the fosdenopterin molecule.

We are obligated to use commercially reasonable efforts to obtain a PRV, achieve specified milestone events and commercialize at least one product containing the fosdenopterin molecule after receipt of regulatory approval.

BBP-009 (Patidegib): Option Agreement with LEO Pharma A/S

In November 2018, through PellePharm, we entered into an option agreement with LEO Pharma, or LEO Pharma, and LEO Spiny Merger Sub, Inc., pursuant to which LEO Pharma was granted an exclusive, irrevocable option to acquire PellePharm. The option is exercisable by LEO Pharma on or before the occurrence of certain events relating to PellePharm’s clinical development programs, and in no event later than July 30, 2021. As consideration for the option, LEO Pharma paid to PellePharm exclusivity payments totaling approximately $27.9 million in the aggregate and purchased a minority equity interest in PellePharm for approximately $5.1 million. In addition, LEO Pharma has agreed to pay additional exclusivity payments to PellePharm in an amount not to exceed $37.0 million in the aggregate under certain circumstances.

Pursuant to the option agreement, we have agreed to conduct the business of PellePharm in the ordinary course and in accordance with applicable laws, comply with the terms of our organizational documents, and use commercially reasonable efforts to operate the business of PellePharm in accordance with a mutually agreed budget and to complete a Phase 2 clinical trial of patidegib for HF-BCC and a Phase 3 clinical trial of patidegib for Gorlin Syndrome. In addition, we and LEO Pharma have formed a joint development committee to oversee the development of, and to make decisions regarding the commercialization of, patidegib.

BBP-589: Asset Purchase Agreement with Shire Human Genetic Therapies, Inc. and Lotus Tissue Repair

In July 2017, through our subsidiary, Phoenix Tissue Repair, Inc. or Phoenix, we entered into an asset purchase agreement with Shire Human Genetic Therapies, Inc., or Shire, and Lotus Tissue Repair, Inc. or Lotus, pursuant to which we acquired from Shire and Lotus the right, title and interest in certain assets relating to recombinant human collagen type VII, including patents and other intellectual property rights, as well as data and regulatory filings, relating to the treatment of DEB, and assumed certain liabilities with respect thereto. In connection with the acquisition of such assets, (i) Shire and Lotus granted to us a non-exclusive, worldwide, irrevocable, perpetual, royalty-free, sublicensable license under certain intellectual property related to the acquired assets but retained by Shire and Lotus, for the exploitation of certain recombinant human collagen type VII products in all fields, and (ii) we granted to Shire and Lotus a non-exclusive, worldwide, irrevocable, perpetual, royalty-free, sublicensable license under certain of the acquired intellectual property assets to exploit products other than recombinant human collagen type VII products and other than for the treatment of DEB in humans.

As partial consideration for our acquisition of the assets, we agreed to pay a purchase price of $1.5 million and issued shares of common stock in Phoenix at a nominal value to Lotus. We are obligated to make contingent milestone payments totaling $27.0 million upon achievement of certain regulatory milestones. In addition, we are obligated to make contingent milestone payments totaling $60.0 million upon achievement of certain sales milestones. We also agreed to pay to Shire and Lotus tiered single-digit royalties on annual net sales for products containing the recombinant human collagen type VII.

We are obligated to use commercially reasonable efforts to develop, obtain FDA approval for and commercialize at least one product for the treatment of DEB in humans.

24


BBP-454: License Agreement with Regents of The University of California

In September 2016, through our subsidiary TheRas, Inc., or TheRas, we entered into a license agreement with the Regents of the University of California, or UCSF, which was amended in January 2017, August 2017, September 2018 and December 2019, relating to certain patent rights related to KRAS inhibitors and modulators, which we refer to collectively as the UCSF License.

Under the UCSF License, we acquired an exclusive, royalty-bearing, sublicensable, worldwide license to make, have made, use, sell, offer for sale and import products, services and methods covered by the licensed patent rights, and to perform licensed processes, in each case, in prophylactic and therapeutic uses in humans. In addition, we received an option for certain inventions conceived and reduced to practice during a specified term. Under the UCSF License, UCSF retains, on behalf of itself and a third party, the right to make, use and practice certain of the licensed intellectual property rights for research and educational purposes, and the right to license to other academic and nonprofit organizations to practice the patent rights for research and educational purposes, including with respect to sponsored research performed on behalf of commercial entities. The rights and interests of any such commercial entity shall be subject to the licenses granted to us pursuant to the UCSF License. The UCSF License is also subject to pre-existing rights of the U.S. Government and the NIH.

In connection with the UCSF License and subsequent amendments, we paid issue fees totaling $300,000. In addition, under the terms of the UCSF License, we are required to pay to UCSF certain annual license maintenance fees unless we are selling or otherwise exploiting licensed products or services paying royalties to UCSF on net sales for such licensed products or services. With respect to such royalty obligations, we agreed to pay UCSF low single-digit tiered royalties on annual net sales of licensed products and services, with a minimum royalty requirement of $100,000. Our obligation to pay royalties continues on a country-by-country basis until the expiration of all licensed patent rights covering licensed products in such country. In addition, we are obligated to make contingent milestone payments totaling up to $22.4 million upon the achievement of certain clinical or regulatory milestones. In the event that we sublicense the patent rights, UCSF is also entitled to receive a percentage of the sublicensing income received by us.

We are also required to make a one-time “Index Milestone Payment” to UCSF in the event of (i) an initial public offering, or (ii) a change of control transaction, in each case with respect to TheRas. Such Index Milestone Payment is calculated by multiplying (a) a number of shares equal to a specified percentage of the then-outstanding fully-diluted shares of common stock of TheRas by (b)(1) in the case of an initial public offering by TheRas, the offering price per share of the securities sold to the underwriters in the offering, or (2) in the case of a change of control transaction with respect to TheRas, the per share consideration that would be received by TheRas’ shareholders in such transaction, in each case subject to certain adjustments. To the extent that an Index Milestone Payment becomes due prior to a bona fide financing transaction of at least $45 million, such Index Milestone Payment is equal to the greater of the amount calculated as described above, or $1.8 million.

Under the UCSF License, we also assumed certain obligations with respect to fund-raising, and must report on our progress in achieving the milestones set forth in the UCSF License on a periodic basis. The UCSF License also includes certain participation rights pursuant to which UCSF has the right to purchase specified amounts of securities offered by TheRas in financing transactions.

Under the UCSF License, we are obligated to diligently proceed with the development, manufacture and sale of at least one licensed product and/or service, and to earnestly and diligently market such licensed product and/or service after receipt of any requisite regulatory approvals and in quantities sufficient to meet market demand. We are also required to use good faith and diligent efforts to meet the milestones set forth in the UCSF License, subject to any revisions that may be permitted under certain circumstances. UCSF has the right to either terminate the UCSF License or reduce the license to a nonexclusive license if we are unable to perform our diligence obligations.

The agreement will continue until the last to expire or abandonment of the patent rights on a licensed product-by-licensed product and country-by-country basis. We may terminate the agreement by providing prior written notice to UCSF or we may terminate the rights under patent rights on a country-by-country basis by giving notice in writing to UCSF. UCSF has the right to terminate the agreement if we fail to make any payments, challenge any UCSF patent rights or otherwise materially breach the agreement and fail to cure such breach within a specified grace period.

25


BBP-398: Collaboration and License Agreement with the Board of Regents of The University of Texas System and The University of Texas M.D. Anderson Cancer Center

In March 2017, through our subsidiary Navire Pharma, Inc. (formerly known as PTP Pharmaceuticals, Inc.), or Navire, we entered into a collaboration and license agreement with The Board of Regents of the University of Texas System, or the Board of Regents, and The University of Texas M.D. Anderson Cancer Center, or MD Anderson. Under the agreement, we acquired an exclusive, royalty-bearing, sublicensable, worldwide license to develop, make, use and sell SHP2 and PTPN11 inhibitors covered by the licensed technology in all fields. The Board of Regents and MD Anderson each retain the right to practice the licensed patent rights for non-commercial, research and academic purposes, and also to grant non-exclusive licenses to other academic and nonprofit organizations to practice the patent rights for non-commercial, research and educational purposes (but excluding any research sponsored by a for-profit entity). Our license is also subject to a non-exclusive license granted to the U.S. government. To further the goals of the collaboration agreement, we granted a non-exclusive license to our technology to MD Anderson for the purpose of carrying out the development plan.

In partial consideration for the exclusive license grant, we issued the Board of Regents shares of common stock of Navire valued at approximately $280,000 pursuant to a stock purchase agreement entered into simultaneously. If commercial sales of a licensed product commence, we will pay MD Anderson royalties at percentage rates ranging in the low single digits on net sales of licensed products. We may offset payments made to third parties to obtain rights needed for the commercialization of a licensed product against royalties payable to MD Anderson provided that such expenses in a given year may not be credited against more than a specified percentage of the royalties in such year and subject to a minimum floor in the low single digits. Our obligation to pay various royalties continues on a country by country basis with respect to any licensed product depends on regulatory status, patent coverage, and financing status. For licensed products that satisfy certain regulatory conditions, the related royalty extends for three years after the first sale. Additionally, if certain financing conditions are achieved, then (i) for licensed products covered by licensed patents, the royalty obligation continues until the expiration of all licensed patent rights covering such licensed product in such country, and (ii) for licensed products without coverage by licensed patents, the royalty obligation extends for 10 years after first sale.

Under the collaboration and license agreement, we are obligated to use commercially reasonable efforts to conduct all development activities under the agreement and to commercialize the licensed products following regulatory approval.

The agreement will continue for thirty years unless earlier terminated. We may terminate the agreement for convenience, provided that MD Anderson shall not be required to forego payments made or equity issued to MD Anderson under the collaboration and license agreement or the stock purchase agreement. MD Anderson has the right to terminate the agreement if we fail to pay royalties or otherwise materially breach the agreement or the stock purchase agreement and fail to cure such breach within a specified cure period, or if BridgeBio Pharma LLC commits a material breach of its obligations under any agreement with Navire, or if Navire breaches obligations under a Series A Preferred Stock Purchase Agreement between Navire and BridgeBio Pharma LLC.

Government Regulation

Government authorities in the United States at the federal, state and local level and in other countries regulate, among other things, the research, development, manufacture, testing, quality control, approval, labeling, packaging, storage, record-keeping, promotion, advertising, distribution, post-approval monitoring and reporting, marketing and export and import of drug and biological products, including gene therapies, as well as diagnostics, and any future product candidates. Generally, before a new drug, biologic or diagnostic can be marketed, considerable data demonstrating its quality, safety and efficacy must be obtained, organized into a format specific for each regulatory authority, submitted for review and approved, authorized, or cleared by the applicable regulatory authority.

U.S. Government Regulation of Drug and Biological Products

In the United States, the FDA regulates drugs under the Federal Food, Drug and Cosmetic Act, or FDCA, and its implementing regulations and biologics under the FDCA and the Public Health Service Act, or PHSA, and their implementing regulations. Both drugs and biologics also are subject to other federal, state and local statutes and regulations, such as those related to competition. The process of obtaining regulatory approvals and the subsequent

26


compliance with appropriate federal, state, and local statutes and regulations requires the expenditure of substantial time and financial resources. Failure to comply with the applicable U.S. requirements at any time during the product development process, approval process or following approval may subject an applicant to administrative actions or judicial sanctions. These actions and sanctions could include, among other actions, the FDA’s refusal to approve pending applications, withdrawal of an approval, license revocation, a clinical hold, untitled or warning letters, voluntary or mandatory product recalls or market withdrawals, product seizures, total or partial suspension of production or distribution, injunctions, fines, refusals of government contracts, restitution, disgorgement and civil or criminal fines or penalties. Any agency or judicial enforcement action could have a material adverse effect on our business, the market acceptance of our products and our reputation.

Our product candidates must be approved by the FDA through either an NDA or a Biologics License Application, or BLA, process before they may be legally marketed in the United States. The process generally involves the following:

 

completion of extensive preclinical studies in accordance with applicable regulations, including studies conducted in accordance with Good Laboratory Practices, or GLP, requirements;

 

submission to the FDA of an IND, which must become effective before human clinical trials may begin;

 

approval by an Institutional Review Board, or IRB, or independent ethics committee at each clinical trial site before each human trial may be initiated;

 

performance of adequate and well-controlled human clinical trials in accordance with applicable IND regulations, Good Clinical Practices, or GCP, requirements and other clinical trial-related regulations to establish the safety and efficacy of the investigational product for each proposed indication;

 

submission to the FDA of an NDA or BLA;

 

a determination by the FDA within 60 days of its receipt of an NDA or BLA to accept the filing for review;

 

satisfactory completion of one or more FDA pre-approval inspections of the manufacturing facility or facilities where the drug or biologic will be produced to assess compliance with Current Good Manufacturing Practices, or cGMP, requirements to assure that the facilities, methods and controls are adequate to preserve the drug or biologic’s identity, strength, quality and purity;

 

potential FDA audit of the clinical trial sites that generated the data in support of the NDA or BLA;

 

payment of user fees for FDA review of the NDA or BLA; and

 

FDA review and approval of the NDA or BLA, including consideration of the views of any FDA advisory committee, prior to any commercial marketing or sale of the drug or biologic in the United States.

The preclinical and clinical testing and approval process requires substantial time, effort and financial resources, and the regulatory scheme for drugs and biologics is evolving and subject to change at any time. We cannot be certain that any approvals for our product candidates will be granted on a timely basis, or at all.

Preclinical Studies

Before testing any drug, biological or gene therapy candidate in humans, the product candidate must undergo rigorous preclinical testing. Preclinical studies include laboratory evaluation of product chemistry and formulation, as well as in vitro and animal studies to assess safety and in some cases to establish a rationale for therapeutic use. The conduct of preclinical studies is subject to federal and state regulations and requirements, including GLP regulations for safety/toxicology studies.

27


An IND sponsor must submit the results of the preclinical tests, together with manufacturing information, analytical data, any available clinical data or literature and plans for clinical trials, among other things, to the FDA as part of an IND. An IND is a request for authorization from the FDA to administer an investigational product to humans, and must become effective before human clinical trials may begin. Some long-term preclinical testing, such as animal tests of reproductive AEs and carcinogenicity, may continue after the IND is submitted. An IND automatically becomes effective 30 days after receipt by the FDA, unless before that time, the FDA raises concerns or questions related to one or more proposed clinical trials and places the trial on clinical hold. In such a case, the IND sponsor and the FDA must resolve any outstanding concerns before the clinical trial can begin. As a result, submission of an IND may not result in the FDA allowing clinical trials to commence. Additionally, the review of information in an IND submission may prompt FDA to, among other things, scrutinize existing INDs or marketed products and could generate requests for information or clinical holds on other product candidates or programs.

Clinical Trials

The clinical stage of development involves the administration of the investigational product to healthy volunteers or patients under the supervision of qualified investigators, generally physicians not employed by or under the trial sponsor’s control, in accordance with GCP requirements, which include the requirement that all research subjects provide their informed consent for their participation in any clinical trial. Clinical trials are conducted under protocols detailing, among other things, the objectives of the clinical trial, dosing procedures, subject selection and exclusion criteria and the parameters to be used to monitor subject safety and assess efficacy. Each protocol, and any subsequent amendments to the protocol, must be submitted to the FDA as part of the IND. Furthermore, each clinical trial must be reviewed and approved by an IRB for each institution at which the clinical trial will be conducted to ensure that the risks to individuals participating in the clinical trials are minimized and are reasonable in relation to anticipated benefits. The IRB also approves the informed consent form that must be provided to each clinical trial subject or his or her legal representative, and must monitor the clinical trial until completed. There also are requirements governing the reporting of ongoing clinical trials and completed clinical trial results to public registries. Information about certain clinical trials, including clinical trial results, must be submitted within specific timeframes for publication on the www.clinicaltrials.gov website.

In addition to the submission of an IND to the FDA before initiation of a clinical trial in the United States, certain human clinical trials involving recombinant or synthetic nucleic acid molecules had historically been subject to review by the NIH Recombinant DNA Advisory Committee, or the RAC, of the NIH Office of Biotechnology Activities, or the OBA, pursuant to the NIH Guideline. On August 17, 2018, the NIH issued a notice in the Federal Register and issued a public statement proposing changes to the oversight framework for gene therapy trials, including changes to the applicable NIH Guidelines to modify the roles and responsibilities of the RAC with respect to human clinical trials of gene therapy products, and requesting public comment on its proposed modifications. During the public comment period, which closed October 16, 2018, the NIH announced that it will no longer accept new human gene transfer protocols for review as a part of the protocol registration process or convene the RAC to review individual clinical protocols. In April 2019, NIH announced the updated guidelines, which reflect these proposed changes, and clarify that these trials will remain subject to the FDA’s oversight and other clinical trial regulations, and oversight at the local level will continue as set forth in the NIH Guidelines. Specifically, under the NIH Guidelines, supervision of human gene transfer trials includes evaluation and assessment by an Institutional Biosafety Committee, or an IBC, a local institutional committee that reviews and oversees research utilizing recombinant or synthetic nucleic acid molecules at that institution. The IBC assesses the safety of the research and identifies any potential risk to public health or the environment, and such review may result in some delay before initiation of a clinical trial. While the NIH Guidelines are not mandatory unless the research in question being conducted at or sponsored by institutions receiving NIH funding of recombinant or synthetic nucleic acid molecule research, many companies and other institutions not otherwise subject to the NIH Guidelines voluntarily follow them.

A sponsor who wishes to conduct a clinical trial outside of the United States may, but need not, obtain FDA authorization to conduct the clinical trial under an IND. If a foreign clinical trial is not conducted under an IND, the sponsor may submit data from the clinical trial to the FDA in support of an NDA or BLA. The FDA will accept a well-designed and well-conducted foreign clinical study not conducted under an IND if the study was conducted in accordance with GCP requirements, and the FDA is able to validate the data through an onsite inspection if deemed necessary.

28


Clinical trials generally are conducted in three sequential phases, known as Phase 1, Phase 2 and Phase 3, and may overlap.

 

Phase 1 clinical trials generally involve a small number of healthy volunteers or disease-affected patients who are initially exposed to a single dose and then multiple doses of the product candidate. The primary purpose of these clinical trials is to assess the metabolism, pharmacologic action, side effect tolerability and safety of the product candidate.

 

Phase 2 clinical trials involve studies in disease-affected patients to evaluate proof of concept and/or determine the dose required to produce the desired benefits. At the same time, safety and further PK and PD information is collected, possible adverse effects and safety risks are identified, and a preliminary evaluation of efficacy is conducted.

 

 

Phase 3 clinical trials generally involve a large number of patients at multiple sites and are designed to provide the data necessary to demonstrate the effectiveness of the product for its intended use, its safety in use and to establish the overall benefit/risk relationship of the product and provide an adequate basis for product labeling.

 

In August 2018, the FDA released a draft guidance entitled “Expansion Cohorts: Use in First-In-Human Clinical Trials to Expedite Development of Oncology Drugs and Biologics,” which outlines how drug developers can utilize an adaptive trial design commonly referred to as a seamless trial design in early stages of oncology drug development, i.e., the first-in-human clinical trial, to compress the traditional three phases of trials into one continuous trial called an expansion cohort trial. Information to support the design of individual expansion cohorts are included in IND applications and assessed by FDA. Expansion cohort trials can potentially bring efficiency to drug development and reduce developmental costs and time.

Post-approval trials, sometimes referred to as Phase 4 clinical trials, may be conducted after initial marketing approval. These trials are used to gain additional experience from the treatment of patients in the intended therapeutic indication and are commonly intended to generate additional safety data regarding use of the product in a clinical setting. In certain instances, the FDA may mandate the performance of Phase 4 clinical trials as a condition of approval of an NDA or BLA.

Progress reports detailing the results of the clinical trials, among other information, must be submitted at least annually to the FDA and written IND safety reports must be submitted to the FDA and the investigators 15 days after the trial sponsor determines the information qualifies for reporting for serious and unexpected suspected AEs, findings from other studies or animal or in vitro testing that suggest a significant risk for human subjects and any clinically important increase in the rate of a serious suspected adverse reaction over that listed in the protocol or investigator brochure. The sponsor must also notify the FDA of any unexpected fatal or life-threatening suspected adverse reaction as soon as possible but in no case later than seven calendar days after the sponsor’s initial receipt of the information.

Phase 1, Phase 2, Phase 3 and other types of clinical trials may not be completed successfully within any specified period, if at all. The FDA or the sponsor may suspend or terminate a clinical trial at any time on various grounds, including a finding that the research subjects or patients are being exposed to an unacceptable health risk. Similarly, an IRB can suspend or terminate approval of a clinical trial at its institution if the clinical trial is not being conducted in accordance with the IRB’s requirements or if the drug or biologic has been associated with unexpected serious harm to patients. Additionally, some clinical trials are overseen by an independent group of qualified experts organized by the clinical trial sponsor, known as a data safety monitoring board or committee. This group provides authorization for whether a trial may move forward at designated check points based on access to certain data from the trial. Concurrent with clinical trials, companies usually complete additional animal studies and also must develop additional information about the chemistry and physical characteristics of the drug or biologic as well as 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 and, among other things, companies must develop methods for testing the identity, strength, quality and purity of the final product. Additionally, appropriate packaging must be selected and tested, and stability studies must be conducted to demonstrate that the product candidates do not undergo unacceptable deterioration over their shelf life.

29


FDA Review Process

Following completion of the clinical trials, data are analyzed to assess whether the investigational product is safe and effective for the proposed indicated use or uses. The results of preclinical studies and clinical trials are then submitted to the FDA as part of an NDA or BLA, along with proposed labeling, chemistry and manufacturing information to ensure product quality and other relevant data. The NDA or BLA is a request for approval to market the drug or biologic for one or more specified indications and must contain proof of safety and efficacy for a drug or safety, purity and potency for a biologic. The application may include both negative and ambiguous results of preclinical studies and clinical trials, as well as positive findings. Data may come from company-sponsored clinical trials intended to test the safety and efficacy of a product’s use or from a number of alternative sources, including studies initiated by investigators. To support marketing approval, the data submitted must be sufficient in quality and quantity to establish the safety and efficacy of the investigational product to the satisfaction of FDA. FDA approval of an NDA or BLA must be obtained before a drug or biologic may be marketed in the United States.

Under the Prescription Drug User Fee Act, or PDUFA, as amended, each NDA or BLA must be accompanied by a user fee. FDA adjusts the PDUFA user fees on an annual basis. Fee waivers or reductions are available in certain circumstances, including a waiver of the application fee for the first application filed by a small business. Additionally, no user fees are assessed on NDAs or BLAs for products designated as orphan drugs, unless the product also includes a non-orphan indication.

The FDA reviews all submitted NDAs and BLAs before it accepts them for filing, and may request additional information rather than accepting the NDA or BLA for filing. The FDA must make a decision on accepting an NDA or BLA for filing within 60 days of receipt, and such decision could include a refusal to file by the FDA. Once the submission is accepted for filing, the FDA begins an in-depth review of the NDA or BLA. Under the goals and policies agreed to by the FDA under PDUFA, the FDA targets ten months, from the filing date, in which to complete its initial review of a new molecular entity NDA or original BLA and respond to the applicant, and six months from the filing date of a new molecular entity NDA or original BLA designated for priority review. The FDA does not always meet its PDUFA goal dates for standard and priority NDAs or BLAs, and the review process is often extended by FDA requests for additional information or clarification.

The FDA has developed the Oncology Center of Excellence RTOR pilot program to facilitate a more efficient review process for certain oncology product candidates. Although this program allows FDA to begin reviewing clinical data prior to submission of a complete NDA or BLA, the program is not intended to change the PDUFA review timelines.

Before approving an NDA or BLA, the FDA will conduct a pre-approval inspection of the manufacturing facilities for the new product to determine whether they comply with cGMP requirements. The FDA will not approve the product 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. The FDA also may audit data from clinical trials to ensure compliance with GCP requirements. Additionally, the FDA may refer applications for novel products or products which present difficult questions of safety or efficacy to an advisory committee, typically a panel that includes clinicians and other experts, for review, evaluation and a recommendation as to whether the application should be approved and under what conditions, if any. The FDA is not bound by recommendations of an advisory committee, but it considers such recommendations when making decisions on approval. The FDA likely will reanalyze the clinical trial data, which could result in extensive discussions between the FDA and the applicant during the review process. After the FDA evaluates an NDA or BLA, it will issue an approval letter or a Complete Response Letter. An approval letter authorizes commercial marketing of the drug or biologic with specific prescribing information for specific indications. A Complete Response Letter indicates that the review cycle of the application is complete, and the application will not be approved in its present form. A Complete Response Letter usually describes all of the specific deficiencies in the NDA or BLA identified by the FDA. The Complete Response Letter may require the applicant to obtain additional clinical data, including the potential requirement to conduct additional pivotal Phase 3 clinical trial(s) and/or to complete other significant and time-consuming requirements related to clinical trials, or to conduct additional preclinical studies or manufacturing activities. If a Complete Response Letter is issued, the applicant may either resubmit the NDA or BLA, addressing all of the deficiencies identified in the letter, or withdraw the application or request an opportunity for a hearing. Even if such data and information are submitted, the FDA may decide that the

30


NDA or BLA does not satisfy the criteria for approval. Data obtained from clinical trials are not always conclusive and the FDA may interpret data differently than we interpret the same data.

Orphan Drug Designation and Exclusivity

Under the Orphan Drug Act, the FDA may grant orphan designation to a drug or biological product intended to treat a rare disease or condition, which is generally 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 and for which there is no reasonable expectation that the cost of developing and making the product available in the United States for this type of disease or condition will be recovered from sales of the product.

Orphan drug designation must be requested before submitting an NDA or BLA. After the FDA grants orphan drug designation, the identity of the therapeutic agent and its potential orphan use are disclosed publicly by the FDA. Orphan drug designation does not convey any advantage in or shorten the duration of the regulatory review and approval process.

If a product that has orphan drug designation subsequently receives the first FDA approval for the disease or condition 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 to market the same drug for the same indication for seven years from the date of such approval, except in limited circumstances, such as a showing of clinical superiority to the product with orphan exclusivity by means of greater effectiveness, greater safety or providing a major contribution to patient care or in instances of drug supply issues. Competitors, however, may receive approval of either a different product for the same indication or the same product for a different indication but that could be used off-label in the orphan indication. Orphan drug exclusivity also could block the approval of one of our products for seven years if a competitor obtains approval before we do for the same product, as defined by the FDA, for the same indication we are seeking approval, or if our product is determined to be contained within the scope of the competitor’s product for the same indication or disease. If we pursue marketing approval for an indication broader than the orphan drug designation we have received, we may not be entitled to orphan drug exclusivity. Orphan drug status in the European Union has similar, but not identical, requirements and benefits.

Rare Pediatric Disease Designation and Priority Review Vouchers

Under the FDCA, as amended, the FDA incentivizes the development of drugs and biologics that meet the definition of a “rare pediatric disease,” defined to mean a serious or life-threatening disease in which the serious of life-threatening manifestations primarily affect individuals aged from birth to 18 years and the disease affects fewer than 200,000 individuals in the United States or affects more than 200,000 in the United States and for which there is no reasonable expectation that the cost of developing and making in the United States a drug for such disease or condition will be received from sales in the United States of such drug. The sponsor of a product candidate for a rare pediatric disease may be eligible for a voucher that can be used to obtain a priority review for a subsequent human drug or biologic application after the date of approval of the rare pediatric disease drug product, referred to as a priority review voucher, or PRV. A sponsor may request rare pediatric disease designation from the FDA prior to the submission of its NDA or BLA. A rare pediatric disease designation does not guarantee that a sponsor will receive a PRV upon approval of its NDA or BLA. Moreover, a sponsor who chooses not to submit a rare pediatric disease designation request may nonetheless receive a PRV upon approval of their marketing application if they request such a voucher in their original marketing application and meet all of the eligibility criteria. If a PRV is received, it may be sold or transferred an unlimited number of times. Congress has extended the PRV program through September 30, 2024, with the potential for PRVs to be granted through September 30, 2026.

Expedited Development and Review Programs

A sponsor may seek to develop and obtain approval of its product candidates under programs designed to accelerate the development, FDA review and approval of new drugs and biologics that meet certain criteria. For example, the FDA has a fast track program that is intended to expedite or facilitate the process for reviewing new drugs and biologics that are intended to treat a serious or life threatening disease or condition and demonstrate the potential to address unmet medical needs for the condition. Fast track designation applies to both the product and the specific indication for which it is being studied. For a fast track-designated product, the FDA may consider sections

31


of the NDA or BLA for review on a rolling basis before the complete application is submitted, if the sponsor provides a schedule for the submission of the sections of the application, the FDA agrees to accept sections of the application and determines that the schedule is acceptable, and the sponsor pays any required user fees upon submission of the first section of the application. The sponsor can request the FDA to designate the product for fast track status any time before receiving NDA or BLA approval, but ideally no later than the pre-NDA or pre-BLA meeting.

A product submitted to the FDA for marketing, including under a fast track program, may be eligible for other types of FDA programs intended to expedite development or review, such as priority review and accelerated approval. Priority review means that, for a new molecular entity or original BLA, the FDA sets a target date for FDA action on the marketing application at six months after accepting the application for filing as opposed to ten months. A product is eligible for priority review if it is designed to treat a serious or life-threatening disease condition and, if approved, would provide a significant improvement in safety and effectiveness compared to available therapies. The FDA will attempt to direct additional resources to the evaluation of an application for a new drug or biologic designated for priority review in an effort to facilitate the review. If criteria are not met for priority review, the application for a new molecular entity or original BLA is subject to the standard FDA review period of ten months after FDA accepts the application for filing. Priority review designation does not change the scientific/medical standard for approval or the quality of evidence necessary to support approval.

A product may also be eligible for accelerated approval if it is designed to treat a serious or life-threatening disease or condition and demonstrates an effect on either 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, or IMM, that is reasonably likely to predict an effect on IMM or other clinical benefit, taking into account the severity, rarity, or prevalence of the disease or condition and the availability or lack of alternative treatments. As a condition of approval, the FDA may require that a sponsor of a drug or biologic receiving accelerated approval perform adequate and well-controlled post-marketing clinical trials. 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. FDA may withdraw approval of a drug or indication approved under accelerated approval if, for example, the confirmatory trial fails to verify the predicted clinical benefit of the product.

Additionally, a drug or biologic may be eligible for designation as a breakthrough therapy if the product is intended, alone or in combination with one or more other drugs or biologics, to treat a serious or life-threatening condition and preliminary clinical evidence indicates that the product may demonstrate substantial improvement over currently approved therapies on one or more clinically significant endpoints, such as substantial treatment effects observed early in clinical development. If the FDA designates a breakthrough therapy, it may take actions appropriate to expedite the development and review of the application, which may include holding meetings with the sponsor and the review team throughout the development of the therapy; providing timely advice to, and interactive communication with, the sponsor regarding the development of the drug to ensure that the development program to gather the nonclinical and clinical data necessary for approval is as efficient as practicable; involving senior managers and experienced review staff, as appropriate, in a collaborative, cross-disciplinary review; assigning a cross-disciplinary project lead for the FDA review team to facilitate an efficient review of the development program and to serve as a scientific liaison between the review team and the sponsor; and considering alternative clinical trial designs when scientifically appropriate, which may result in smaller trials or more efficient trials that require less time to complete and may minimize the number of patients exposed to a potentially less efficacious treatment. Breakthrough therapy designation comes with all of the benefits of fast track designation, which means that the sponsor may file sections of the BLA for review on a rolling basis if certain conditions are satisfied, including an agreement with the FDA on the proposed schedule for submission of portions of the application and the payment of applicable user fees before the FDA may initiate a review.

As part of the 21st Century Cures Act, Congress amended the FDCA to facilitate an efficient development program for, and expedite review of regenerative medicine advanced therapies, or RMATs, which include cell and gene therapies, therapeutic tissue engineering products, human cell and tissue products, and combination products using any such therapies or products. RMATs do not include those human cells, tissues, and cellular and tissue based products regulated solely under section 361 of the PHSA and 21 CFR Part 1271. This program is intended to facilitate efficient development and expedite review of regenerative medicine therapies, which are intended to treat, modify, reverse, or cure a serious or life-threatening disease or condition and qualify for RMAT designation. A drug

32


sponsor may request that the FDA designate a drug as a RMAT concurrently with or at any time after submission of an IND. The FDA has 60 calendar days to determine whether the drug meets the criteria, including whether there is preliminary clinical evidence indicating that the drug has the potential to address unmet medical needs for a serious or life-threatening disease or condition. A BLA for a regenerative medicine therapy that has received RMAT designation may be eligible for priority review or accelerated approval through use of surrogate or intermediate endpoints reasonably likely to predict long-term clinical benefit, or reliance upon data obtained from a meaningful number of sites. Benefits of RMAT designation also include early interactions with FDA to discuss any potential surrogate or intermediate endpoint to be used to support accelerated approval. A regenerative medicine therapy with RMAT designation that is granted accelerated approval and is subject to post-approval requirements may fulfill such requirements through the submission of clinical evidence from clinical studies, patient registries, or other sources of real world evidence, such as electronic health records; the collection of larger confirmatory data sets; or post-approval monitoring of all patients treated with such therapy prior to its approval.

The FDA has also announced the availability of the RTOR pilot program for oncology product candidates that are likely to demonstrate substantial improvements over available therapy, which may include drugs previously granted breakthrough therapy designation for the same or other indications and candidates meeting other criteria for other expedited programs, such as fast track and priority review. Submissions for RTOR consideration should also have straightforward study designs and endpoints that can be easily interpreted (such as overall survival or progression free survival). Acceptance into the RTOR pilot does not guarantee or influence approvability of the application, which is subject to the usual benefit-risk evaluation by FDA reviewers, but the program allows FDA to review data earlier, before an applicant formally submits a complete application. The RTOR pilot program does not affect FDA’s PDUFA timelines.

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 the time period for FDA review or approval may not be shortened. Furthermore, fast track designation, priority review, accelerated approval, breakthrough therapy and RMAT designation do not change the standards for approval.

Pediatric Information and Pediatric Exclusivity

Under the Pediatric Research Equity Act, or PREA, certain NDAs and BLAs and certain supplements to an NDA or BLA must contain data to assess the safety and efficacy of the drug 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 FDA may grant deferrals for submission of pediatric data or full or partial waivers. The Food and Drug Administration Safety and Innovation Act, or FDASIA, amended the FDCA to require that a sponsor who is planning to submit a marketing application for a drug 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, or PSP, within 60 days of an end-of-Phase 2 meeting or, if there is no such meeting, as early as practicable before the initiation of the Phase 3 or Phase 2/3 study. The initial PSP must include an outline of the pediatric study or studies that the sponsor plans to conduct, including study objectives and design, age groups, relevant endpoints and statistical approach, or a justification for not including such detailed information, and any request for a deferral of pediatric assessments or a full or partial waiver of the requirement to provide data from pediatric studies along with supporting information. The FDA and the sponsor must reach an agreement on the PSP. A sponsor can submit amendments to an agreed-upon initial PSP at any time if changes to the pediatric plan need to be considered based on data collected from preclinical studies, early phase clinical trials and/or other clinical development programs.

A drug or biologic 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.

Post-Marketing Requirements

Following approval of a new product, the manufacturer and the approved product are subject to continuing regulation by the FDA, including, among other things, monitoring and record-keeping activities, reporting of

33


adverse experiences, complying with promotion and advertising requirements, which include restrictions on promoting products for unapproved uses or patient populations (known as “off-label use”) and limitations on industry-sponsored scientific and educational activities. Although physicians may prescribe legally available products for off-label uses, manufacturers may not market or promote such uses. The FDA and other agencies actively enforce the laws and regulations prohibiting the promotion of off-label uses, and a company that is found to have improperly promoted off-label uses may be subject to significant liability, including investigation by federal and state authorities. Prescription drug promotional materials must be submitted to the FDA in conjunction with their first use or first publication. Further, if there are any modifications to the drug or biologic, including changes in indications, labeling or manufacturing processes or facilities, the applicant may be required to submit and obtain FDA approval of a new NDA/BLA or NDA/BLA supplement, which may require the development of additional data or preclinical studies and clinical trials.

The FDA may also place other conditions on approvals including the requirement for a REMS, to assure the safe use of the product. If the FDA concludes a REMS is needed, the sponsor of the NDA or BLA must submit a proposed REMS. The FDA will not approve the NDA or BLA without an approved REMS, if required. A REMS 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. Any of these limitations on approval or marketing could restrict the commercial promotion, distribution, prescription or dispensing of products. Product approvals may be withdrawn for non-compliance with regulatory standards or if problems occur following initial marketing.

FDA regulations require that products be manufactured in specific approved facilities and in accordance with cGMP regulations. We rely, and expect to continue to rely, on third parties for the production of clinical and commercial quantities of our products in accordance with cGMP regulations. These manufacturers must comply with cGMP regulations that require, among other things, quality control and quality assurance, the maintenance of records and documentation and the obligation to investigate and correct any deviations from cGMP. Manufacturers and other entities involved in the manufacture and distribution of approved drugs or biologics 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 cGMP requirements and other laws. Accordingly, manufacturers must continue to expend time, money and effort in the area of production and quality control to maintain cGMP compliance. The discovery of violative conditions, including failure to conform to cGMP regulations, could result in enforcement actions, and the discovery of problems with a product after approval may result in restrictions on a product, manufacturer or holder of an approved NDA or BLA, including recall.

Once an approval is granted, the FDA may issue enforcement letters or withdraw the approval of the product if compliance with regulatory requirements and standards is not maintained or if problems occur after the drug or biologic reaches the market. Corrective action could delay drug or biologic distribution and require significant time and financial expenditures. Later discovery of previously unknown problems with a drug or biologic, including AEs 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 trials to assess new safety risks; or imposition of distribution or other restrictions under a REMS program. Other potential consequences include, among other things:

 

restrictions on the marketing or manufacturing of the drug or biologic, suspension of the approval, complete withdrawal of the drug from the market or product recalls;

 

 

fines, warning letters or holds on post-approval clinical trials;

 

refusal of the FDA to approve applications or supplements to approved applications, or suspension or revocation of drug or biologic approvals;

 

 

drug or biologic seizure or detention, or refusal to permit the import or export of drugs; or

 

injunctions or the imposition of civil or criminal penalties.

34


Regulation of Companion Diagnostics

We believe that the success of certain of our product candidates may depend, in part, on the development and commercialization of a companion diagnostic. Companion diagnostics identify patients who are most likely to benefit from a particular therapeutic product; identify patients likely to be at increased risk for serious side effects as a result of treatment with a particular therapeutic product; or monitor response to treatment with a particular therapeutic product for the purpose of adjusting treatment to achieve improved safety or effectiveness. Companion diagnostics are regulated as medical devices by the FDA. 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 or FDA exercise of enforcement discretion applies, diagnostic tests generally require marketing clearance or approval from the FDA prior to commercialization. The two primary types of FDA marketing authorization applicable to a medical device are premarket notification, also called 510(k) clearance, and approval of a premarket, or PMA approval.

To obtain 510(k) clearance for a medical device, or for certain modifications to devices that have received 510(k) clearance, a manufacturer must submit a premarket notification demonstrating that the proposed device is substantially equivalent to a previously cleared 510(k) device or to a preamendment device that was in commercial distribution before May 28, 1976, or a predicate device, for which the FDA has not yet called for the submission of a PMA. In making a determination that the device is substantially equivalent to a predicate device, the FDA compares the proposed device to the predicate device or predicate devices and assesses whether the subject device is comparable to the predicate device or predicate devices with respect to intended use, technology, design and other features which could affect safety and effectiveness. If the FDA determines that the subject device is substantially equivalent to the predicate device or predicate devices, the subject device may be cleared for marketing. The 510(k) premarket notification pathway generally takes from three to twelve months from the date the application is completed, but can take significantly longer.

PMA applications must be supported by valid scientific evidence, which typically requires extensive data, including technical, preclinical, clinical and manufacturing data, to demonstrate to the FDA’s satisfaction the safety and effectiveness of the device. For diagnostic tests, a PMA application typically includes data regarding analytical and clinical validation studies. As part of its review of the PMA, the FDA will conduct a pre-approval inspection of the manufacturing facility or facilities to ensure compliance with the Quality System Regulation, or QSR, which requires manufacturers to follow design, testing, control, documentation and other quality assurance procedures. The FDA’s review of an initial PMA application is required by statute to take between six to ten months, although the process typically takes longer, and may require several years to complete. If the FDA evaluations of both the PMA application and the manufacturing facilities are favorable, the FDA will either issue an approval letter or an approvable letter, which usually contains a number of conditions that must be met in order to secure the final approval of the PMA. If the FDA’s evaluation of the PMA or manufacturing facilities is not favorable, the FDA will deny the 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. Once granted, PMA approval may be withdrawn by the FDA if compliance with post-approval requirements, conditions of approval or other regulatory standards is not maintained or problems are identified following initial marketing.

On July 31, 2014, the FDA issued a final guidance document addressing the development and approval process for “In Vitro Companion Diagnostic Devices.” According to the guidance document, for novel therapeutic products that depend on the use of a diagnostic test and where the diagnostic device could be essential for the safe and effective use of the corresponding therapeutic product, the premarket application for the companion diagnostic device should be developed and approved or cleared contemporaneously with the therapeutic, although the FDA recognizes that there may be cases when contemporaneous development may not be possible. However, in cases where a drug cannot be used safely or effectively without the companion diagnostic, the FDA’s guidance indicates it will generally not approve the drug without the approval or clearance of the diagnostic device. The FDA also issued a draft guidance in July 2016 setting forth the principles for co-development of an in vitro companion diagnostic device with a therapeutic product. The draft guidance describes principles to guide the development and contemporaneous marketing authorization for the therapeutic product and its corresponding in vitro companion diagnostic.

35


Once cleared or approved, the companion diagnostic device must adhere to post-marketing requirements including the requirements of FDA’s quality system regulation, adverse event reporting, recalls and corrections along with product marketing requirements and limitations. Like drug and biologic makers, companion diagnostic makers are subject to unannounced FDA inspections at any time during which the FDA will conduct an audit of the product(s) and the company’s facilities for compliance with its authorities.

Biosimilars and Exclusivity

Certain of our product candidates are regulated as biologics. An abbreviated approval pathway for biological products shown to be similar to, or interchangeable with, an FDA-licensed reference biological product was created by the Biologics Price Competition and Innovation Act of 2009, or BPCI Act, as part of the ACA. This amendment to the PHSA, in part, attempts to minimize duplicative testing. Biosimilarity, which requires that the biological product be highly similar to the reference product notwithstanding minor differences in clinically inactive components and that there be no clinically meaningful differences between the product and the reference product in terms of safety, purity and potency, can be shown through analytical studies, animal studies and a clinical trial or trials. Interchangeability requires that a biological product be biosimilar to the reference product and that the product can be expected to produce the same clinical results as the reference product in any given patient and, for products administered multiple times to an individual, that the product and the reference product 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 biological product without such alternation or switch. Complexities associated with the larger, and often more complex, structure of biological products as compared to small molecule drugs, as well as the processes by which such products are manufactured, pose significant hurdles to implementation that are still being worked out by the FDA.

A reference biological product is granted four and twelve year exclusivity periods from the time of first licensure of the product. The FDA will not accept an application for a biosimilar or interchangeable product based on the reference biological product until four years after the date of first licensure of the reference product, and the FDA will not approve an application for a biosimilar or interchangeable product based on the reference biological product until twelve years after the date of first licensure of the reference product. “First licensure” typically means the initial date the particular product at issue was licensed in the United States. Date of first licensure does not include the date of licensure of (and a new period of exclusivity is not available for) a biological product if the licensure is for a supplement for the biological product or for a subsequent application by the same sponsor or manufacturer of the biological product (or licensor, predecessor in interest, or other related entity) for a change (not including a modification to the structure of the biological product) that results in a new indication, route of administration, dosing schedule, dosage form, delivery system, delivery device or strength, or for a modification to the structure of the biological product that does not result in a change in safety, purity, or potency. Therefore, one must determine whether a new product includes a modification to the structure of a previously licensed product that results in a change in safety, purity, or potency to assess whether the licensure of the new product is a first licensure that triggers its own period of exclusivity. Whether a subsequent application, if approved, warrants exclusivity as the “first licensure” of a biological product is determined on a case-by-case basis with data submitted by the sponsor.

Other Regulatory Matters

Manufacturing, sales, promotion and other activities following product approval are also subject to regulation by numerous regulatory authorities in the United States in addition to the FDA, including the Centers for Medicare and Medicaid Services, or CMS, including the Office of Inspector General and Office for Civil Rights, other divisions of the Department of HHS, the Department of Justice, the Drug Enforcement Administration, the Consumer Product Safety Commission, the Federal Trade Commission, the Occupational Safety & Health Administration, the Environmental Protection Agency and state and local governments.

Healthcare providers, physicians, and third party payors will play a primary role in the recommendation and prescription of any products for which we obtain marketing approval. Our current and future arrangements with healthcare providers and physicians and any future arrangements with third party payers, may expose us to broadly applicable fraud and abuse and other healthcare laws and regulations that may constrain the business or financial arrangements and relationships through which we market, sell and distribute any drugs for which we obtain marketing approval. In the United States, these laws include: the federal Anti-Kickback Statute, the False Claims Act, and HIPAA.

36


The Anti-Kickback Statute makes it illegal for any person, including a prescription drug manufacturer (or a party acting on its behalf), to knowingly and willfully solicit, receive, offer or pay any remuneration, directly or indirectly, in cash or in kind, that is intended to induce or reward referrals, including the purchase, recommendation, order or prescription of a particular drug, for which payment may be made under a federal healthcare program, such as Medicare or Medicaid. Violations of this law are punishable by imprisonment, criminal fines, administrative civil money penalties and exclusion from participation in federal healthcare programs. In addition, a person or entity does not need to have actual knowledge of the statute or specific intent to violate it. Moreover, the ACA provides that 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 federal civil False Claims Act.

Although we would not submit claims directly to payors, drug manufacturers can be held liable under the federal civil False Claims Act, which imposes civil penalties, including through civil whistleblower or qui tam actions, against individuals or entities (including manufacturers) for, among other things, knowingly presenting, or causing to be presented to federal programs (including Medicare and Medicaid) claims for items or services, including drugs, that are false or fraudulent, claims for items or services not provided as claimed, or claims for medically unnecessary items or services. Penalties for a False Claims Act violation include three times the actual damages sustained by the government, plus mandatory civil penalties for each separate false claim, the potential for exclusion from participation in federal healthcare programs and, although the federal False Claims Act is a civil statute, conduct that results in a False Claims Act violation may also implicate various federal criminal statutes. The government may deem manufacturers to have “caused” the submission of false or fraudulent claims by, for example, providing inaccurate billing or coding information to customers or promoting a product off-label. Claims which include items or services resulting from a violation of the federal Anti-Kickback Statute are false or fraudulent claims for purposes of the False Claims Act. Our future marketing and activities relating to the reporting of wholesaler or estimated retail prices for our products, if approved, the reporting of prices used to calculate Medicaid rebate information and other information affecting federal, state and third-party reimbursement for our products, and the sale and marketing of our product candidates, are subject to scrutiny under this law.

HIPAA created federal criminal statutes that prohibit among other things, knowingly and willfully executing, or attempting to execute, a scheme to defraud or to obtain, by means of false or fraudulent pretenses, representations or promises, any money or property owned by, or under the control or custody of, any healthcare benefit program, including private third party payors, knowingly and willfully embezzling or stealing from a healthcare benefit program, willfully obstructing a criminal investigation of a healthcare offense, and knowingly and willfully falsifying, concealing or covering up by trick, scheme or device, a material fact or making any materially false, fictitious or fraudulent statement in connection with the delivery of or payment for healthcare benefits, items or services. Like 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 civil monetary penalties statute imposes penalties against any person or entity that, among other things, is determined to have presented or caused to be presented a claim to a federal health program that the person knows or should know is for an item or service that was not provided as claimed or is false or fraudulent.

We may be subject to data privacy and security regulations by both the federal government and the states in which we conduct our business. HIPAA, as amended by HITECH, and their implementing regulations, mandates, among other things, the adoption of uniform standards for the electronic exchange of information in common healthcare transactions, as well as standards relating to the privacy and security of individually identifiable health information, which require the adoption of administrative, physical and technical safeguards to protect such information. Among other things, HITECH makes HIPAA’s security standards directly applicable to business associates, defined as independent contractors or agents of covered entities, which include certain health care providers, health plans, and healthcare clearinghouses, that create, receive or obtain protected health information in connection with providing a service for or on behalf of a covered entity. HITECH also increased the civil and criminal penalties that may be imposed against covered entities and business associates, and gave state attorneys general new authority to file civil actions for damages or injunctions in federal courts to enforce the federal HIPAA laws and seek attorney’s fees and costs associated with pursuing federal civil actions. In addition, certain state laws govern the privacy and security of health information in certain circumstances, some of which are more stringent than HIPAA and many of which differ from each other in significant ways and may not have the same effect, thus complicating compliance efforts. Failure to comply with these laws, where applicable, can result in the imposition of significant civil and criminal penalties.

37


Additionally, the federal Physician Payments Sunshine Act, or the Sunshine Act, within the ACA, and its implementing regulations, require that certain manufacturers of drugs, devices, biological and medical supplies for which payment is available under Medicare, Medicaid or the Children’s Health Insurance Program (with certain exceptions) report annually to CMS information related to certain payments or other transfers of value made or distributed to physicians and teaching hospitals, or to entities or individuals at the request of, or designated on behalf of, physicians, certain other healthcare professionals, and teaching hospitals and to report annually certain ownership and investment interests held by physicians, certain other healthcare professionals, and their immediate family members. Effective January 1, 2022, these reporting obligations will extend to include transfers of value made to certain non-physician providers such as physician assistants and nurse practitioners. In addition, many states also govern the reporting of payments or other transfers of value, many of which differ from each other in significant ways, are often not pre-empted, and may have a more prohibitive effect than the Sunshine Act, thus further complicating compliance efforts.

Similar federal, state and foreign fraud and abuse 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. Such laws are generally broad and are enforced by various state agencies and private actions. Also, many states have similar fraud and abuse statutes or regulations that may be broader in scope and may apply regardless of payor, in addition to items and services reimbursed under Medicaid and other state programs. Some state laws require pharmaceutical companies to comply with the pharmaceutical industry’s voluntary compliance guidelines and the relevant federal government compliance guidance, and require drug manufacturers to report information related to payments and other transfers of value to physicians and other healthcare providers or marketing expenditures.

In order to distribute products commercially, we must comply with state laws that require the registration of manufacturers and wholesale distributors of drug and biological products in a state, including, in certain states, manufacturers and distributors who ship products into the state even if such manufacturers or distributors have no place of business within the state. Several states have enacted legislation requiring pharmaceutical and biotechnology companies to establish marketing compliance programs, file periodic reports with the state, make periodic public disclosures on sales, marketing, pricing, clinical trials and other activities, and/or register their sales representatives, as well as to prohibit pharmacies and other healthcare entities from providing certain physician prescribing data to pharmaceutical and biotechnology companies for use in sales and marketing, and to prohibit certain other sales and marketing practices. All of our activities are potentially subject to federal and state consumer protection and unfair competition laws.

The scope and enforcement of each of these laws is uncertain and subject to rapid change in the current environment of healthcare reform, especially in light of the lack of applicable precedent and regulations. Federal and state enforcement bodies have recently increased their scrutiny of interactions between healthcare companies and healthcare providers, which has led to a number of investigations, prosecutions, convictions and settlements in the healthcare industry. 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 civil, criminal and administrative penalties, damages, fines, disgorgement, contractual damages, reputational harm, diminished profits and future earnings, individual imprisonment, exclusion of drugs from government funded healthcare programs, such as Medicare and Medicaid, and the curtailment or restructuring of our operations, any of which could adversely affect our ability to operate our business and our financial results. 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 criminal, civil or administrative sanctions, including exclusions from government funded healthcare programs. Ensuring business arrangements comply with applicable healthcare laws, as well as responding to possible investigations by government authorities, can be time- and resource-consuming and can divert a company’s attention from the business.

Current and Future Legislation

In the United States and some foreign jurisdictions, there have been, and likely will continue to be, a number of legislative and regulatory changes and proposed changes regarding the healthcare system directed at broadening the availability of healthcare, improving the quality of healthcare, and containing or lowering the cost of healthcare.

38


For example, in March 2010, the ACA was enacted in the United States. The ACA includes measures that have significantly changed, and are expected to continue to significantly change, the way healthcare is financed by both governmental and private insurers. Among the provisions of the ACA of greatest importance to the pharmaceutical industry are that the ACA:

 

made several changes to the Medicaid Drug Rebate Program, including increasing pharmaceutical manufacturers’ rebate liability by raising the minimum basic Medicaid rebate on most branded prescription drugs to 23.1% of average manufacturer price, or AMP, and adding a new rebate calculation for “line extensions” (i.e., new formulations, such as extended release formulations) of solid oral dosage forms of branded products, as well as potentially impacting their rebate liability by modifying the statutory definition of AMP.

 

imposed a requirement on manufacturers of branded drugs to provide a 70% (increased pursuant to the Bipartisan Budget Act of 2018, effective as of 2019) point-of-sale discount off the negotiated price of branded drugs dispensed to Medicare Part D beneficiaries in the coverage gap (i.e., “donut hole”) as a condition for a manufacturer’s outpatient drugs being covered under Medicare Part D.

 

extended a manufacturer’s Medicaid rebate liability to covered drugs dispensed to individuals who are enrolled in Medicaid managed care organizations.

 

expanded the entities eligible for discounts under the 340B Drug Discount Program.

 

established a new methodology by which rebates owed by manufacturers under the Medicaid Drug Rebate Program are calculated for drugs that are inhaled, infused, instilled, implanted, or injected.

 

imposed an annual, nondeductible fee on any entity that manufactures or imports certain branded prescription drugs, apportioned among these entities according to their market share in certain government healthcare programs.

 

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. The research conducted by the Patient-Centered Outcomes Research Institute may affect the market for certain pharmaceutical products. The ACA established the Center for Medicare and Medicaid Innovation within 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 numerous judicial, administrative, executive, and legislative challenges to certain aspects of the ACA, and we expect there will be additional challenges and amendments to the ACA in the future. Various portions of the ACA are currently undergoing legal and constitutional challenges in the United States Supreme Court; the Trump Administration issued various Executive Orders which eliminated cost sharing subsidies and various provisions that would impose a fiscal burden on states or a cost, fee, tax, penalty or regulatory burden on individuals, healthcare providers, health insurers, or manufacturers of pharmaceuticals or medical devices; and Congress has introduced several pieces of legislation aimed at significantly revising or repealing the ACA. The United States Supreme Court is expected to rule on a legal challenge to the constitutionality of the ACA in early 2021. The implementation of the ACA is ongoing, the law appears likely to continue the downward pressure on pharmaceutical pricing, especially under the Medicare program.

Moreover, in May 2018, the Trump administration released its “Blueprint to Lower Drug Prices and Reduce Out-of-Pocket Costs,” or the Blueprint. The Blueprint contains several potential regulatory actions and legislative recommendations aimed at lowering prescription drug prices, including measures to promote innovation and competition for biologics, changes to Medicare Part D to give plan sponsors more leverage when negotiating prices with manufacturers, and updating the Medicare drug-pricing dashboard to make price increases and generic competition more transparent. HHS has already implemented certain of these measures, while others are pending. For example, in May 2019, the CMS issued a final rule to allow Medicare Advantage Plans the option of using step therapy, a type of prior authorization, for Part B drugs beginning January 1, 2020. Additional recommendations in

39


the Blueprint and RFI, if enacted by Congress and HHS, could lead to changes to Medicare Parts B and D, including the transition of certain drugs covered under Part B to Part D or the offering of alternative purchasing options under the Competitive Acquisition Program that currently applies to selected drugs and biologics covered under Part B. For example, in May 2019, CMS issued a final rule to allow Medicare Advantage Plans the option of using step therapy, a type of prior authorization, for Part B drugs beginning January 1, 2020. This final rule codified CMS’s policy change that was effective January 1, 2019. Although some proposals related to the previous administration’s Blueprint may require additional authorization to become effective, may ultimately be withdrawn, or may face challenges in the courts, Congress has indicated that it will continue to seek new legislative, administrative and/or additional measures to control drug costs. Likewise, the Biden administration has indicated that lowering prescription drug prices is a priority, but we do not yet know what steps the administration will take or whether such steps will be successful.

Other legislative changes have been proposed and adopted in the United States since the ACA was enacted. In August 2011, the Budget Control Act of 2011, among other things, created measures for spending reductions by Congress. A Joint Select Committee on Deficit Reduction, tasked with recommending a targeted deficit reduction of at least $1.2 trillion for the years 2013 through 2021, was unable to reach required goals, thereby triggering the legislation’s automatic reduction to several government programs. This includes aggregate reductions of Medicare payments to providers up to 2% per fiscal year, which went into effect in April 2013, following passage of the Bipartisan Budget Act of 2013, and will remain in effect through 2029 unless additional congressional action is taken. Pursuant to the CARES Act, as well as subsequent legislation, these reductions have been suspended from May 1, 2020 through March 31, 2021 due to the COVID-19 pandemic. Proposed legislation, if passed, would extend this suspension until the end of the pandemic. Additionally, there has been increasing legislative and enforcement interest in the United States with respect to specialty drug pricing practices.

Specifically, there have been several recent U.S. Congressional inquiries and proposed bills designed to, among other things, bring more transparency to drug pricing, review the relationship between pricing and manufacturer patient programs and reform government program reimbursement methodologies for drugs.

Further, on May 30, 2018, the Trickett Wendler, Frank Mongiello, Jordan McLinn, and Matthew Bellina Right to Try Act of 2017, or the Right to Try Act, was signed into law. The law, among other things, provides a federal framework for certain patients to request access to certain investigational new drug products that have completed a Phase I clinical trial and that are undergoing investigation for FDA approval. There is no obligation for a pharmaceutical manufacturer to make its drug products available to eligible patients as a result of the Right to Try Act.

We cannot predict what healthcare reform initiatives may be adopted in the future. Further federal, state and foreign legislative and regulatory developments are likely, and we expect ongoing initiatives to increase pressure on drug pricing. Such reforms could have an adverse effect on anticipated revenues from product candidates and may affect our overall financial condition and ability to develop product candidates.

Packaging and Distribution in the United States

If our products are made available to authorized users of the Federal Supply Schedule of the General Services Administration, additional laws and requirements apply. Products must meet applicable child-resistant packaging requirements under the U.S. Poison Prevention Packaging Act. Manufacturing, sales, promotion and other activities also are potentially subject to federal and state consumer protection and unfair competition laws.

The distribution of pharmaceutical products is subject to additional requirements and regulations, including extensive record-keeping, licensing, storage and security requirements intended to prevent the unauthorized sale of pharmaceutical products.

The failure to comply with any of these laws or regulatory requirements subjects firms to possible legal or regulatory action. Depending on the circumstances, failure to meet applicable regulatory requirements can result in criminal prosecution, fines or other penalties, injunctions, exclusion from federal healthcare programs, requests for recall, seizure of products, total or partial suspension of production, denial or withdrawal of product approvals, or refusal to allow a firm to enter into supply contracts, including government contracts. Any action against us for

40


violation of these laws, even if we successfully defend against it, could cause us to incur significant legal expenses and divert our management’s attention from the operation of our business. Prohibitions or restrictions on sales or withdrawal of future products marketed by us could materially affect our business in an adverse way.

Changes in regulations, statutes or the interpretation of existing regulations could impact our business in the future by requiring, for example: (i) changes to our manufacturing arrangements; (ii) additions or modifications to product labeling; (iii) the recall or discontinuation of our products; or (iv) additional record-keeping requirements. If any such changes were to be imposed, they could adversely affect the operation of our business.

Other U.S. Environmental, Health and Safety Laws and Regulations

We may be subject to numerous environmental, health and safety laws and regulations, including those governing laboratory procedures and the handling, use, storage, treatment and disposal of hazardous materials and wastes. From time to time and in the future, our operations may involve the use of hazardous and flammable materials, including chemicals and biological materials, and may also produce hazardous waste products. Even if we contract with third parties for the disposal of these materials and waste products, we cannot completely eliminate the risk of contamination or injury resulting from these materials. In the event of contamination or injury resulting from the use or disposal of our hazardous materials, we could be held liable for any resulting damages, and any liability could exceed our resources. We also could incur significant costs associated with civil or criminal fines and penalties for failure to comply with such laws and regulations.

We maintain workers’ compensation insurance to cover us for costs and expenses we may incur due to injuries to our employees, but this insurance may not provide adequate coverage against potential liabilities. However, we do not maintain insurance for environmental liability or toxic tort claims that may be asserted against us.

In addition, we may incur substantial costs in order to comply with current or future environmental, health and safety laws and regulations. Current or future environmental laws and regulations may impair our research, development or production efforts. In addition, failure to comply with these laws and regulations may result in substantial fines, penalties or other sanctions.

U.S. Patent Term Restoration and Marketing Exclusivity

Depending upon the timing, duration and specifics of FDA approval of our future product candidates, some of our U.S. patents may be eligible for limited patent term extension under the Drug Price Competition and Patent Term Restoration Act of 1984, commonly referred to as the Hatch-Waxman Amendments. The Hatch-Waxman Amendments permit restoration of the patent term of up to five years as compensation for patent term lost during the FDA regulatory review process. Patent-term restoration, however, cannot extend the remaining term of a patent beyond a total of 14 years from the product’s approval date and only those claims covering such approved drug product, a method for using it or a method for manufacturing it may be extended. The patent-term restoration period is generally one-half the time between the effective date of an IND and the submission date of an NDA or BLA plus the time between the submission date of an NDA or BLA and the approval of that application, except that the review period is reduced by any time during which the applicant failed to exercise due diligence. Only one patent applicable to an approved drug is eligible for the extension and the application for the extension must be submitted prior to the expiration of the patent. The USPTO, in consultation with the FDA, reviews and approves the application for any patent term extension or restoration. In the future, we may apply for restoration of patent term for our currently owned or licensed patents to add patent life beyond its current expiration date, depending on the expected length of the clinical trials and other factors involved in the filing of the relevant NDA or BLA.

Marketing exclusivity provisions under the FDCA also can delay the submission or the approval of certain applications. The FDCA provides a five-year period of non-patent marketing exclusivity within the United States to the first applicant to gain 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 accept for review an ANDA, or a 505(b)(2) NDA submitted by another company for another version of such drug 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. The FDCA also

41


provides three years of marketing exclusivity for an NDA, 505(b)(2) 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 conditions of use associated with the new clinical investigations and does not prohibit the FDA from approving ANDAs for drugs containing the original active agent. 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 all of the preclinical studies and adequate and well-controlled clinical trials necessary to demonstrate safety and effectiveness.

European Union Drug Development

In the European Union, our future products also may be subject to extensive regulatory requirements. As in the United States, medicinal products can be marketed only if a marketing authorization from the competent regulatory agencies has been obtained.

Similar to the United States, the various phases of preclinical and clinical research in the European Union are subject to significant regulatory controls. Although the EU Clinical Trials Directive 2001/20/EC, or Directive, has sought to harmonize the EU clinical trials regulatory framework, setting out common rules for the control and authorization of clinical trials in the EU, the EU Member States have transposed and applied the provisions of the Directive differently into their national laws. This has led to significant variations in the Member State regimes. Under the current regime, before a clinical trial can be initiated it must be approved in each of the EU countries where the trial is to be conducted by two distinct bodies: the National Competent Authority, or NCA, and one or more Ethics Committees, or ECs. Under the current regime all suspected unexpected serious adverse reactions to the investigated drug that occur during the clinical trial have to be reported to the NCA and ECs of the Member State where they occurred.

The EU clinical trials legislation currently is undergoing a transition process mainly aimed at harmonizing and streamlining clinical-trial authorization, simplifying adverse-event reporting procedures, improving the supervision of clinical trials and increasing their transparency. In April 2014, the EU adopted a new Clinical Trials Regulation (EU) No 536/2014, or Regulation, which is set to replace the current Clinical Trials Directive 2001/20/EC. It will overhaul the current system of approvals for clinical trials in the EU. Specifically, the new Regulation, which will be directly applicable in all Member States, aims at simplifying and streamlining the approval of clinical trials in the EU. For instance, the new Regulation provides for a streamlined application procedure via a single entry point and strictly defined deadlines for the assessment of clinical trial applications. It is expected that the new Regulation will apply following confirmation of full functionality of the Clinical Trials Information System, the centralized EU portal and database for clinical trials foreseen by the regulation, through an independent audit, which is currently anticipated to occur in December 2021.

European Union Drug Marketing

Much like the Anti-Kickback Statue prohibition in the United States, the provision of benefits or advantages to physicians to induce or encourage the prescription, recommendation, endorsement, purchase, supply, order or use of medicinal products is also prohibited in the European Union. The provision of benefits or advantages to induce or reward improper performance generally is governed by the national anti-bribery laws of the European Union Member States, and the Bribery Act 2010 in the UK. Infringement of these laws could result in substantial fines and imprisonment. EU Directive 2001/83/EC, which is the EU Directive governing medicinal products for human use, further provides that, where medicinal products are being promoted to persons qualified to prescribe or supply them, no gifts, pecuniary advantages or benefits in kind may be supplied, offered or promised to such persons unless they are inexpensive and relevant to the practice of medicine or pharmacy. This provision has been transposed into the Human Medicines Regulations 2012 and so remains applicable in the UK despite its departure from the EU.

Payments made to physicians in certain EU Member States must be publicly disclosed. Moreover, agreements with physicians often must be the subject of prior notification and approval by the physician’s employer, his or her competent professional organization and/or the regulatory authorities of the individual EU Member States. These requirements are provided in the national laws, industry codes or professional codes of conduct, applicable in the EU

42


Member States. Failure to comply with these requirements could result in reputational risk, public reprimands, administrative penalties, fines or imprisonment.

European Union Drug Review and Approval

In the European Economic Area, or EEA, which is comprised of the Member States of the European Union plus Norway, Iceland and Liechtenstein, medicinal products can only be commercialized after obtaining a marketing authorization, or MA. There are two types of marketing authorizations.

 

The centralized MA is issued by the European Commission through the centralized procedure, based on the opinion of the Committee for Medicinal Products for Human Use, or CHMP, of the EMA, and is valid throughout the entire territory of the EEA. The centralized procedure is mandatory for certain types of products, such as biotechnology medicinal products, orphan medicinal products, advanced-therapy medicinal products (gene-therapy, somatic cell-therapy or tissue-engineered medicines) and medicinal products containing a new active substance indicated for the treatment of HIV, AIDS, cancer, neurodegenerative disorders, diabetes, auto-immune and other immune dysfunctions and viral diseases. The centralized procedure is optional for products containing a new active substance not yet authorized in the EEA, or for products that constitute a significant therapeutic, scientific or technical innovation or which are in the interest of public health in the EEA.

Under the centralized procedure the maximum timeframe for the evaluation of a marketing authorization application, or MAA, by the EMA is 210 days, excluding clock stops, when additional written or oral information is to be provided by the applicant in response to questions asked by the CHMP. Clock stops may extend the timeframe of evaluation of a MAA considerably beyond 210 days. Where the CHMP gives a positive opinion, it provides the opinion together with supporting documentation to the European Commission, who make the final decision to grant a marketing authorization, which is issued within 67 days of receipt of the EMA’s recommendation. Accelerated assessment might be granted by the CHMP in exceptional cases, when a medicinal product is expected to be of major public health interest, particularly from the point of view of therapeutic innovation. The timeframe for the evaluation of a MAA under the accelerated assessment procedure is 150 days, excluding clock stops, but it is possible that the CHMP may revert to the standard time limit for the centralized procedure if it determines that the application is no longer appropriate to conduct an accelerated assessment.

 

National MAs, which are issued by the competent authorities of the Member States of the EEA 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 a Member State of the EEA, this national MA can be recognized in another Member States 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, or RMS. The competent authority of the RMS prepares a draft assessment report, a draft summary of the product characteristics, or SmPC, and a draft of the labeling and package leaflet, which are sent to the other Member States (referred to as the Concerned Member States) for their approval. If the Concerned Member States raise no objections, based on a potential serious risk to public health, to the assessment, SmPC, labeling, or packaging proposed by the RMS, the product is subsequently granted a national MA in all the Member States (i.e., in the RMS and the Concerned Member States).

 

Under the above described procedures, before granting the MA, the EMA or the competent authorities of the Member States of the EEA make an assessment of the risk-benefit balance of the product on the basis of scientific criteria concerning its quality, safety and efficacy.

Now that the United Kingdom (which comprises Great Britain and Northern Ireland) has left the European Union, Great Britain will no longer be covered by centralized MAs (under the Northern Irish Protocol, centralized MAs will continue to be recognized in Northern Ireland). All medicinal products with a current centralized MA

43


were automatically converted to Great Britain MAs on January, 1 2021. For a period of two years from January 1, 2021, the Medicines and Healthcare products Regulatory Agency, or MHRA, the UK medicines regulator, may rely on a decision taken by the European Commission on the approval of a new MA in the centralized procedure, in order to more quickly grant a new Great Britain MA. A separate application will, however, still be required. The MHRA also has the power to have regard to MAs approved in EEA Member States through decentralized or mutual recognition procedures with a view to more quickly granting a MA in the United Kingdom or Great Britain.

European Union New Chemical Entity Exclusivity

In the EEA, innovative medicinal products qualify for eight years of data exclusivity upon marketing authorization and an additional two years of market exclusivity. The data exclusivity, if granted, prevents generic or biosimilar applicants from referencing the innovator’s pre-clinical and clinical trial data contained in the dossier of the reference product when applying for a generic or biosimilar MA in the EEA, for a period of eight years from the date of authorization of the reference product. During the additional two-year period of market exclusivity, a generic marketing authorization application can be submitted, and the innovator’s data may be referenced, but no generic or biosimilar product can be marketed until the expiration of the market exclusivity. The overall ten-year period will be extended to a maximum of 11 years if, during the first eight years of those ten years, the marketing authorization holder obtains an authorization for one or more new therapeutic indications which, during the scientific evaluation prior to their authorization, are determined to bring a significant clinical benefit in comparison with currently approved therapies. Even if an innovative medicinal product gains the prescribed period of data exclusivity, however, another company may market another version of the product if such company obtained a MA based on a marketing authorization application with a complete independent data package of pharmaceutical tests, preclinical tests and clinical trials.

European Union Orphan Designation and Exclusivity

In the EEA, the EMA’s Committee for Orphan Medicinal Products grants orphan drug designation to promote the development of products that are intended for the diagnosis, prevention or treatment of life-threatening or chronically debilitating conditions and either (i) such condition affects not more than five in 10,000 persons in the EEA, or (ii) it is unlikely that the development of the medicine would generate sufficient return to justify the necessary investment in its development. In either case, the applicant must also demonstrate that no satisfactory method of diagnosis, prevention or treatment has been authorized (or, if a method exists, the product would be a significant benefit to those affected compared to the product available).

In the EEA, orphan drug designation entitles a party to financial incentives such as reduction of fees or fee waivers and ten years of market exclusivity is granted following grant of a marketing authorization. During this market exclusivity period, neither the EMA nor the European Commission nor any of the competent authorities in the EEA Members States can accept an application or grant a marketing authorization for a “similar medicinal product.” A “similar medicinal product” is defined as a medicinal product containing a similar active substance or substances as contained in an authorized orphan medicinal product, and which is intended for the same therapeutic indication. This period may be reduced to six years if the orphan drug designation criteria are no longer met, including where it is shown that the product is sufficiently profitable not to justify maintenance of market exclusivity. Market exclusivity may also be revoked in very select cases, such as if (i) it is established that a similar medicinal product is safer, more effective or otherwise clinically superior to the authorized product; (ii) the marketing authorization holder consents to such revocation; or (iii) the marketing authorization holder cannot supply enough orphan medicinal product. Orphan drug designation must be requested before submitting an application for marketing approval. Orphan drug designation does not convey any advantage in, or shorten the duration of, the regulatory review and approval process.

From January 1, 2021, a separate process for orphan drug designation will apply in Great Britain. There will be no pre-marketing authorization orphan designation (as there is in the EEA) and the application for orphan designation will be reviewed by the MHRA at the time of the marketing authorization application. The criteria are the same as in the EEA, save that they apply to Great Britain only (e.g., there must be no satisfactory method of diagnosis, prevention or treatment of the condition concerned in Great Britain).

44


European Pediatric Investigation Plan

In the EEA, MAAs for new medicinal products have to include the results of studies conducted in the pediatric population, in compliance with a pediatric investigation plan, or PIP, agreed with the EMA’s Pediatric Committee, or PDCO. The PIP sets out the timing and measures proposed to generate data to support a pediatric indication of the drug for which marketing authorization 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 are 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 this 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. If a marketing authorization is obtained and trial results are included in the product information, even when negative, the product is eligible for six months’ supplementary protection certificate extension. In the case of orphan medicinal products, a two year extension of the orphan market exclusivity may be available. This pediatric reward is subject to specific conditions and is not automatically available when data in compliance with the PIP are developed and submitted.

Brexit and the Regulatory Framework in the United Kingdom

 

On June 23, 2016, the electorate in the United Kingdom voted in favor of leaving the European Union (commonly referred to as Brexit). Thereafter, on March 29, 2017, the country formally notified the European Union of its intention to withdraw pursuant to Article 50 of the Lisbon Treaty. The United Kingdom formally left the European Union on January 31, 2020. A transition period began on February 1, 2020, during which EU pharmaceutical law remained applicable to the United Kingdom. However this ended on December 31, 2020. Since the regulatory framework in the United Kingdom covering the quality, safety and efficacy of pharmaceutical products, clinical trials, marketing authorizations, commercial sales, and distribution of pharmaceutical products is derived from EU Directives and Regulations, Brexit could materially impact the future regulatory regime which applies to products and the approval of product candidates in the United Kingdom, as the UK legislation now has the potential to diverge from EU legislation. It remains to be seen how Brexit will impact regulatory requirements for medicinal products and devices in the United Kingdom in the long term. The MHRA has recently published detailed guidance for industry and organizations to follow now the transition period is over, which will be updated as the United Kingdom’s regulatory position on medicinal products and medical devices evolves over time.

European Data Collection

The collection and use of personal health data in the EEA is governed by the General Data Protection Regulation, or GDPR, which became effective May 25, 2018. The GDPR applies to any company established in the EEA and to companies established outside the EEA that process personal data in connection with the offering of goods or services to data subjects in the European Union or the monitoring of the behavior of data subjects in the European Union. The GDPR enhances data protection obligations for data controllers of personal data, including stringent requirements relating to the consent of data subjects, expanded disclosures about how personal data is used, requirements to conduct privacy impact assessments for “high risk” processing, limitations on retention of personal data, mandatory data breach notification and “privacy by design” requirements, and creates direct obligations on service providers acting as data processors. The GDPR also imposes strict rules on the transfer of personal data outside of the EEA to countries that do not ensure an adequate level of protection, like the United States. Failure to comply with the requirements of the GDPR and the related national data protection laws of the EEA Member States may result in fines up to €20 million or 4% of a company’s global annual revenues for the preceding financial year, whichever is higher. Moreover, the GDPR grants data subjects the right to claim material and non-material damages resulting from infringement of the GDPR. Given the breadth and depth of changes in data protection obligations, maintaining compliance with the GDPR, will require significant time, resources and expense, and we may be required to put in place additional mechanisms ensuring compliance with the new data protection rules. This may be onerous and adversely affect our business, financial condition, results of operations and prospects.

In addition, further to the United Kingdom’s exit from the European Union on January 31, 2020, the GDPR ceased to apply in the United Kingdom at the end of the transition period on December 31, 2020. However, as of

45


January 1, 2021, the United Kingdom’s European Union (Withdrawal) Act 2018 incorporated the GDPR (as it existed on December 31, 2020 but subject to certain UK specific amendments) into UK law, referred to as the UK GDPR. The UK GDPR and the UK Data Protection Act 2018 set out the United Kingdom’s data protection regime, which is independent from but aligned to the European Union’s data protection regime. Non-compliance with the UK GDPR may result in monetary penalties of up to £17.5 million or 4% of worldwide revenue, whichever is higher. The United Kingdom, however, is now regarded as a third country under the European Union’s GDPR which means that transfers of personal data from the EEA to the United Kingdom will be restricted unless an appropriate safeguard, as recognized by the EU’s GDPR, has been put in place. Although, under the EU-UK Trade Cooperation Agreement it is lawful to transfer personal data between the United Kingdom and the EEA for a six-month period following the end of the transition period, with a view to achieving an adequacy decision from the European Commission during that period. Like the EU GDPR, the UK GDPR restricts personal data transfers outside the United Kingdom to countries not regarded by the United Kingdom as providing adequate protection (this means that personal data transfers from the United Kingdom to the EEA remain free flowing).

Rest of the World Regulation

For other countries outside of the European Union and the United States, such as countries in Eastern Europe, Latin America or Asia, the requirements governing the conduct of clinical trials, product licensing, pricing and reimbursement vary from country to country. Additionally, the clinical trials must be conducted in accordance with GCP requirements 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

Additional Laws and Regulations Governing International Operations

If we further expand our operations outside of the United States, we must dedicate additional resources to comply with numerous laws and regulations in each jurisdiction in which we plan to operate. The Foreign Corrupt Practices Act, or FCPA, prohibits any U.S. individual or business from paying, offering, authorizing payment or offering of anything of value, directly or indirectly, to any foreign official, political party or candidate for the purpose of influencing any act or decision of the foreign entity in order to assist the individual or business in obtaining or retaining business. The FCPA also obligates companies whose securities are listed in the United States to comply with certain accounting provisions requiring the company to maintain books and records that accurately and fairly reflect all transactions of the corporation, including international subsidiaries, and to devise and maintain an adequate system of internal accounting controls for international operations.

Compliance with the FCPA is expensive and difficult, particularly in countries in which corruption is a recognized problem. In addition, the FCPA presents particular challenges in the pharmaceutical industry, because, in many countries, hospitals are operated by the government, and doctors and other hospital employees are considered foreign officials. Certain payments to hospitals in connection with clinical trials and other work have been deemed to be improper payments to government officials and have led to FCPA enforcement actions.

Various laws, regulations and executive orders also restrict the use and dissemination outside of the United States, or the sharing with certain non-U.S. nationals, of information classified for national security purposes, as well as certain products and technical data relating to those products. If we expand our presence outside of the United States, it will require us to dedicate additional resources to comply with these laws, and these laws may preclude us from developing, manufacturing, or selling certain products and product candidates outside of the United States, which could limit our growth potential and increase our development costs.

The failure to comply with laws governing international business practices may result in substantial civil and criminal penalties and suspension or debarment from government contracting. The U.S. Securities and Exchange Commission, or the SEC, also may suspend or bar issuers from trading securities on U.S. exchanges for violations of the FCPA’s accounting provisions.

46


Coverage and Reimbursement

Successful commercialization of new drug products depends in part on the extent to which reimbursement for those drug products will be available from government health administration authorities, private health insurers, and other organizations. Government authorities and third-party payors, such as private health insurers and health maintenance organizations, decide which drug products they will pay for and establish reimbursement levels. The availability and extent of reimbursement by governmental and private payors is essential for most patients to be able to afford a drug product. Sales of drug products depend substantially, both domestically and abroad, on the extent to which the costs of drugs products are paid for by health maintenance, managed care, pharmacy benefit and similar healthcare management organizations, or reimbursed by government health administration authorities, private health coverage insurers and other third-party payors.

A primary trend in the U.S. healthcare industry and elsewhere is cost containment. Government authorities and third-party payors have attempted to control costs by limiting coverage and the amount of reimbursement for particular drug products. In many countries, the prices of drug products are subject to varying price control mechanisms as part of national health systems. In general, the prices of drug products under such systems are substantially lower than in the United States. Other countries allow companies to fix their own prices for drug products, but monitor and control company profits. Accordingly, in markets outside the United States, the reimbursement for drug products may be reduced compared with the United States.

In the United States, the principal decisions about reimbursement for new drug products are typically made by CMS, an agency within the HHS. CMS decides whether and to what extent a new drug product will be covered and reimbursed under Medicare, and private payors tend to follow CMS to a substantial degree. However, no uniform policy of coverage and reimbursement for drug products exists among third-party payors and coverage and reimbursement levels for drug products can differ significantly from payor to payor.

Third-party payors may limit coverage to specific products on an approved list or formulary, which might not include all of the FDA-approved products for a particular indication. Also, third-party payors may refuse to include a particular branded drug on their formularies or otherwise restrict patient access to a branded drug when a less costly generic equivalent or another alternative is available. Third-party payors are increasingly challenging the prices charged for medical products and services, examining the medical necessity, and reviewing the cost-effectiveness of medical products and services and imposing controls to manage costs. Further, due to the COVID-19 pandemic, millions of individuals have lost or are expected to lose employer-based insurance coverage, which may adversely affect our ability to successfully commercialize our products.

The Medicare Prescription Drug, Improvement, and Modernization Act of 2003, or the MMA, established the Medicare Part D program to provide a voluntary prescription drug benefit to Medicare beneficiaries. Under Part D, Medicare beneficiaries may enroll in prescription drug plans offered by private entities that provide coverage of outpatient prescription drugs. Unlike Medicare Parts A and B, Part D coverage is not standardized. Part D prescription drug plan sponsors are not required to pay for all covered Part D drugs, and each drug plan can develop its own drug formulary that identifies which drugs it will cover and at what tier or level. While all Medicare drug plans must give at least a standard level of coverage set by Medicare, Part D prescription drug plan sponsors are not required to pay for all covered Part D drugs, and each drug plan can develop its own drug formulary that identifies which drugs it will cover and at what tier or level. However, Part D prescription drug formularies must include drugs within each therapeutic category and class of covered Part D drugs, though not necessarily all the drugs in each category or class. Any formulary used by a Part D prescription drug plan must be developed and reviewed by a pharmacy and therapeutic committee. Government payment for some of the costs of prescription drugs may increase demand for drugs for which we obtain marketing approval. Any negotiated prices for any of our products covered by a Part D prescription drug plan will likely be lower than the prices we might otherwise obtain. Moreover, while the MMA applies only to drug benefits for Medicare beneficiaries, private payors often follow Medicare coverage policy and payment limitations in setting their own payment rates. Any reduction in payment that results from the MMA may result in a similar reduction in payments from non-governmental payors.

For a drug product to receive federal reimbursement under the Medicaid or Medicare Part B programs or to be sold directly to U.S. government agencies, the manufacturer must extend discounts to entities eligible to participate in the 340B drug pricing program. The required 340B discount on a given product is calculated based on the average

47


manufacturer price, or AMP, and Medicaid rebate amounts reported by the manufacturer. As of 2010, the ACA expanded the types of entities eligible to receive discounted 340B pricing, although under the current state of the law these newly eligible entities (with the exception of children’s hospitals) will not be eligible to receive discounted 340B pricing on orphan drugs. As 340B drug pricing is determined based on AMP and Medicaid rebate data, the revisions to the Medicaid rebate formula and AMP definition described above could cause the required 340B discount to increase. The American Recovery and Reinvestment Act of 2009 provides funding for the federal government to compare the effectiveness of different treatments for the same illness. In addition, there have been several changes to the 340B drug pricing program in recent years, which impose ceilings on prices that drug manufacturers can charge for medications sold to certain health care facilities. Legal challenges to reimbursement formula changes under the 340B drug pricing program are ongoing, and it is unclear how these developments could affect covered hospitals who might purchase our future products and affect the rates we may charge such facilities for our approved products in the future, if any.

The plan for the research was published in 2012 by the Department of HHS, the Agency for Healthcare Research and Quality and the National Institutes for Health, and periodic reports on the status of the research and related expenditures are made to Congress. Although the results of the comparative effectiveness studies are not intended to mandate coverage policies for public or private payors, it is not clear what effect, if any, the research will have on the sales of our drug candidates, if any such drug or the condition that they are intended to treat are the subject of a trial. It is also possible that comparative effectiveness research demonstrating benefits in a competitor’s drug could adversely affect the sales of our drug candidate. If third-party payors do not consider our drugs to be cost-effective compared to other available therapies, they may not cover our drugs after approval as a benefit under their plans or, if they do, the level of payment may not be sufficient to allow us to sell our drugs on a profitable basis.

These laws, and future state and federal healthcare reform measures may be adopted in the future, any of which may result in additional reductions in Medicare and other healthcare funding and otherwise affect the prices we may obtain for any product candidates for which we may obtain regulatory approval or the frequency with which any such product candidate is prescribed or used.

Outside of the United States, the pricing of pharmaceutical products and medical devices is subject to governmental control in many countries. For example, in the European Union, pricing and reimbursement schemes vary widely from country to country. Some countries provide that products may be marketed only after a reimbursement price has been agreed. Some countries may require the completion of additional studies that compare the cost effectiveness of a particular therapy to currently available therapies or so-called health technology assessments, in order to obtain reimbursement or pricing approval. Other countries may allow companies to fix their own prices for products, but monitor and control product volumes and issue guidance to physicians to limit prescriptions. Efforts to control prices and utilization of pharmaceutical products and medical devices will likely continue as countries attempt to manage healthcare expenditures.

 

Human Capital Management

 

Our human capital philosophy relies on attracting and retaining team members who consistently demonstrate top performance. Our culture and our approach to talent reinforces this philosophy, including recruiting, professional development, performance management and total rewards. We have provided below additional details on some of our core human resources, or People, processes.

 

As of December 31, 2020, we and our subsidiaries, to which we refer as our affiliates, had 385 full-time employees and 11 part-time employees. Of these, 268 focus on driving forward research and development programs, either directly or through our affiliates, and 128 work across our affiliates to provide strategic business development, finance and executive leadership expertise, as well as general and administrative services generally across our affiliates. We have never had a work stoppage, and none of our employees is represented by a labor organization or under any collective-bargaining arrangements. We consider our employee relations to be good.

 

48


Recruiting

 

In 2020, we established an in-house talent acquisition capability to support our affiliates in hiring the right talent at the right time. This team of experienced recruiters works closely with hiring managers to understand the required skills and capabilities for an open role, and then supports the interview process and evaluation of candidates. We strive to hire top talent, and therefore need a high-quality recruiting process and candidate experience. We endeavor to fill every role with the most qualified candidate possible, which sometimes requires partnership with an external recruitment agency. We are consistently looking at new opportunities and avenues to recruit talented individuals to work at BridgeBio.

 

The talent acquisition team’s focus in 2021 is to meet the growth needs across BridgeBio and our affiliates. We recognize that our current and potential future team members have options for employment opportunities, including with other biotech and pharma companies, research and academic institutions, government entities, and consulting and investment firms. To attract and retain top performing team members, we focus on creating an environment that allows for autonomy, growth and impact while also offering a competitive total rewards package.

 

Professional Development and Performance Management

 

We invest in the professional development of our team members through regular feedback and guidance, as well as targeted learning and development opportunities to meet demonstrated needs. We established a set of five core attributes that we expect every BridgeBio team member to demonstrate while performing in their roles: Patient Champion, Entrepreneurial Operator, Truth Seeker, Inspires Excellence and High-Quality Executor.

 

BridgeBio conducts semi-annual formal performance reviews for all team members to evaluate performance against these attributes. These reviews include self, peer and manager feedback. The feedback focuses on strengths and opportunities for improvement to enable the professional development of all team members. At the end of the year, the performance review also includes a formal rating and informs compensation decisions, including performance bonus, salary adjustments and promotions.

 

Core Values and Ethics

 

Millions worldwide are afflicted with genetic diseases, but small patient populations and industry reluctance to conduct early-stage development means that for many, treatments have not been forthcoming. We are committed to bridging this gap: between business case and scientific possibility, between patient and hope. This starts with our first core value: to put patients first. We also strive to think independently. Our goal is to not simply accept the ideas and opinions of others as fact, but instead to ask “why?” and “why not?” We endeavor to bring a rigorous, first-principles mindset to each problem that we take on. We pride ourselves on being radically transparent. A commitment to independent thinking requires us to consider the ideas of others and to adopt them if they prove best. We strive to maintain a culture where any idea is worthy of both consideration and testing. We know that every minute counts. Our decentralized model strives to deliver treatments from discovery to patients as fast as humanly possible by utilizing focused teams of experts for each asset. Big decisions can be taken by people best-equipped to understand them, without wasting time on unnecessary cycles. And we let Science speak. Our model was designed to promote the rational assessment of our programs. Decisions about a program’s fate are driven by its performance against a set of objective criteria, giving each potential medicine’s scientific merits the last word. All employees are responsible for upholding these values and the BridgeBio Code of Business Conduct and Ethics, which forms the foundation of our policies and practices.

 

Total Rewards

 

To attract and retain top talent, we offer a competitive total rewards package. We peg total direct compensation at the upper end of market. We link a portion of every employee’s compensation to performance through a performance bonus program. To create a sense of ownership and align employee incentives with our long-term success, we offer eligible employees equity ownership in the company through stock option or restricted stock unit grants and our employee stock purchase plan. We also designed a program to incentive affiliate-level employees to achieve specific milestones at core value-inflection points, such as IND or NDA approval.

 

49


We focus our benefits offering on areas critical to keeping our employees and their immediate families healthy and productive. We offer physical and mental health benefits to all employees who work at least 20 hours per week, on average. We have a flexible paid time off policy to empower team members to take the time they need, when they need it.

 

Diversity, Equity and Inclusion

 

We believe that a diverse, equitable and inclusive culture is critical to BridgeBio’s success. We are proud to promote unique voices within and outside our organization, and are eager to learn from others’ experiences, as we know that a diverse and inclusive workforce is a business imperative and key to our long-term success.

 

As a starting point of our Diversity, Equity and Inclusion, or DE&I, efforts, a group of employees launched a “Women at Bridge” employee resource group, which receives direct support from executive management. Women at Bridge is passionate about strengthening opportunities for women and working with others who share enthusiasm for that goal. In its first few months, the group performed an assessment of diversity and inclusion at BridgeBio, identified potential gaps, and formulated steps to address them. They facilitate open and direct communication to engage the broader BridgeBio organization, including updates with our Chief Executive Officer, open forums or town hall meetings, often with a guest speaker, regular ongoing update communications, and employee engagement surveys. Recently, a number of Women at Bridge “chapters” were formed to complement the larger forum and facilitate smaller, more meaningful discussions about strengthening and reinforcing opportunities and engagement of women across the organization.  

 

In 2021, we intend to roll out additional initiatives to guide our DE&I activities. We formed a volunteer DE&I Steering Committee, comprised of a diverse set of BBP employees, to draft a DE&I vision for BridgeBio, and to establish specific goals and priorities for 2021. As one of the DE&I Steering Committee’s first initiatives, we intend to launch an Unconscious Bias education program for employees in 2021.

 

Response to COVID-19

 

With the emergence of the COVID-19 global pandemic, and prior to the shelter-in-place mandate from the State of California where the majority of our employees are located, we took extra precautions to reduce the risk of virus exposure for all employees. We encouraged all employees who were able to work from home to do so. For these newly remote employees, we provided ergonomic evaluations of their workstation, allowed flexible schedules, supported their information technology needs, and provided guidance for managers to ensure that their employees were maintaining their physical, mental and emotional wellbeing. We provided a monthly financial subsidy from April through August to accommodate for any new or ongoing at-home support that was needed.

 

We further supported our employees and government efforts to curb the COVID-19 pandemic through a multifaceted communication, infrastructure, and behavior modification and enforcement effort, including:

 

establishing clear and regular COVID-19 policies, safety protocols, and updates to all employees;

providing/mandating Covid-19 PCR testing protocol for all employees that need or desire to be in the office or lab;

decreasing density and increasing physical distancing in workspaces for employees working onsite by scheduling adjustments;

adjusting attendance policies to encourage those who are sick to stay home;

increasing cleaning protocols across all locations;

providing additional personal protective equipment and cleaning supplies;

implementing protocols to address actual and suspected COVID-19 cases and potential exposure;

50


prohibiting all domestic and international non-essential travel for all employees; and

requiring masks to be worn in all locations.

 

Corporate and Other Information

We were incorporated as a Delaware corporation in 2019, under the name BridgeBio Pharma, Inc. Our principal executive offices are located at 421 Kipling Street, Palo Alto, CA 94301. Our telephone number is (650) 391-9740.

Our web page address is https://bridgebio.com. Our investor relations website is located at https://investor.bridgebio.com. We make available free of charge on our investor relations website under “SEC Filings” our Annual Reports on Form 10‑K, Quarterly Reports on Form 10‑Q, Current Reports on Form 8‑K, our directors’ and officers’ Section 16 Reports and any amendments to those reports after filing or furnishing such materials to the SEC. References to our website address do not constitute incorporation by reference of the information contained on the website, and the information contained on the website is not part of this document or any other document that we file with or furnish to the SEC.

We became a large accelerated filer on December 31, 2020 because our aggregate worldwide market value of the voting and non-voting common equity held by non-affiliates as of June 30, 2020, our most recently completed second fiscal quarter, was greater than $700 million. Prior to that, we were an “emerging growth company”, or EGC, as defined in the Jumpstart Our Business Startups Act of 2012. As an EGC, we were eligible for exemptions from various reporting requirements applicable to other public companies that are not emerging growth companies, including, but not limited to, not being required to comply with the auditor attestation requirements of Section 404 of the Sarbanes-Oxley Act of 2002 and reduced disclosure obligations regarding executive compensation. We can no longer avail ourselves of these exemptions and are now required to comply with the standards and compliance dates for large accelerated filers.

 

51


ITEM 1A.  RISK FACTORS

Our business involves significant risks, some of which are described below. You should carefully consider the risks and uncertainties described below, together with all of the other information contained in this Annual Report on Form 10-K, including “Management’s Discussion and Analysis of Financial Condition and Results of Operations” and the consolidated financial statements and the related notes. If any of the following risks actually occurs, it could harm our business, prospects, operating results and financial condition and future prospects. In such event, the market price of our common stock could decline, and you could lose all or part of your investment. Additional risks and uncertainties not presently known to us or that we currently deem immaterial may also impair our business operations. This Annual Report on Form 10-K also contains forward-looking statements that involve risks and uncertainties. Our actual results could differ materially from those anticipated in the forward-looking statements as a result of factors that are described below and elsewhere in this Annual Report.

Risks Related to Our Financial Position and Growth Strategy

Drug development is a highly uncertain undertaking and involves a substantial degree of risk. We have incurred significant losses since our inception and anticipate that we will continue to incur significant losses for the foreseeable future. We have not generated any revenue since inception, which, together with our limited operating history, may make it difficult for you to assess our future viability.

Pharmaceutical and biopharmaceutical product development is a highly speculative undertaking and involves a substantial degree of risk. We are a clinical-stage biopharmaceutical company with a limited operating history upon which you can evaluate our business and prospects. Our subsidiaries, on whose success we largely rely, are also early-stage biopharmaceutical companies. To date, we have focused principally on identifying, acquiring or in-licensing and developing our product candidates at the subsidiary level, all of which are in discovery, lead optimization, preclinical or clinical development. Our product candidates will require substantial additional development time, including extensive clinical research, and resources before we would be able to apply for or receive regulatory approvals and begin generating revenue from product sales.

We are not profitable and have incurred losses in each year since our inception in April 2015. Our net losses for the years ended December 31, 2020, 2019 and 2018 were $505.5 million, $288.6 million and $169.5 million, respectively. As of December 31, 2020, we had an accumulated deficit of $888.8 million. We have no products approved for commercial sale and have not generated any revenues from product sales, and have financed operations solely through the sale of equity securities and debt financings. We continue to incur significant research and development, or R&D, and other expenses related to ongoing operations and expect to incur losses for the foreseeable future. In addition, we believe that potential delays in our ongoing and planned clinical trials and adjustments to certain of our study procedures, such as enabling alternate site, telehealth and home visits, and at home drug delivery, with respect to our ongoing clinical trials, as a result of the SARS-CoV-2, the novel strain of coronavirus that causes coronavirus disease 19, or COVID-19, pandemic, could increase our expenditures or draw out our expenditures over a longer period of time than originally estimated. Additionally, changes to our selection of contract research organizations, or CROs, for non-clinical laboratory activities and engagement with contract manufacturing organizations, or CMOs, to mitigate any potential near-term impacts to our supply chain may increase our expenditures relative to initial expectations. We anticipate these losses will increase substantially in future periods and we will not generate any revenue from product sales until after we have successfully completed clinical development and received regulatory approval for the commercial sale of one or more product candidates.

Because of the numerous risks and uncertainties associated with drug development, we are unable to predict the timing or amount of our expenses, or when we will be able to generate any meaningful revenue or achieve or maintain profitability, if ever. In addition, our expenses could increase beyond our current expectations if we are required by the U.S. Food and Drug Administration, or FDA, or comparable foreign regulatory authorities, to conduct nonclinical or preclinical studies or clinical trials in addition to those that we currently anticipate or to otherwise provide data beyond that which we currently believe is necessary to support an application for marketing approval or to continue clinical development, or if there are any delays in any of our or our future collaborators’ clinical trials or the development of our product candidates that we may identify. Even if our future product candidates that we may identify are approved for commercial sale, we anticipate incurring significant costs associated with commercializing any approved product candidate and ongoing compliance efforts.

52


We may never be able to develop or commercialize a marketable drug or achieve profitability. Revenue from the sale of any product candidate for which regulatory approval is obtained will be dependent, in part, upon the size of the markets in the territories for which we gain regulatory approval, the accepted price for the product, the ability to obtain reimbursement at any price and whether we own the commercial rights for that territory. Our growth strategy depends on our ability to generate revenue. Even if we achieve profitability in the future, we may not be able to sustain profitability in subsequent periods. Our failure to achieve sustained profitability would depress the value of our company and could impair our ability to raise capital, expand our business, diversify our research and development pipeline, market our product candidates, if approved, that we may identify and pursue, or continue our operations. Our prior losses, combined with expected future losses, have had and will continue to have an adverse effect on our stockholders’ equity and working capital.

If we obtain a controlling interest in additional companies in the future, it could adversely affect our operating results and the value of our common stock, thereby disrupting our business.

As part of our strategy, we expect to form and invest in additional wholly-owned subsidiaries and variable interest entities, or VIEs. Investments in our existing and any future subsidiaries involve numerous risks, including, but not necessarily limited to:

 

risk of conducting research and development activities in new therapeutic areas or treatment modalities in which we have little to no experience;

 

diversion of financial and managerial resources from existing operations;

 

our ability to negotiate a proposed acquisition, in-license or investment in a timely manner or at a price or on terms and conditions favorable to us;

 

our ability to combine and integrate a potential acquisition into our existing business to fully realize the benefits of such acquisition;

 

the impact of regulatory reviews on a proposed acquisition, in-license or investment; and

 

the outcome of any legal proceedings that may be instituted with respect to a potential acquisition, in-license or investment.

 

If we fail to properly evaluate potential acquisitions, in-licenses, investments or other transactions associated with the creation of new research and development programs or the maintenance of existing ones, we might not achieve the anticipated benefits of any such transaction, we might incur costs in excess of what we anticipate, and management resources and attention might be diverted from other necessary or valuable activities. For instance, in January 2021, we completed our acquisition of all of the outstanding shares of common stock of Eidos that were not previously owned by us or our subsidiaries, to which we refer as the Eidos Merger. In connection with the Eidos Merger and our integration of Eidos’ historical operations into our business, the attention of certain members of each company’s management and each company’s resources were and may continue to be diverted from day-to-day business operations, and we may fail to realize the anticipated benefits of the transaction. Additionally, the interests of our stockholders were diluted as a result of our issuance of shares of our common stock to Eidos’ stockholders and our assumption of certain equity awards of Eidos in connection with the transaction. We may engage in similar discussions in the future with respect to other potential transactions that may divert our time and resources from our ongoing operations. In addition, from time to time we have pursued, and may in the future pursue, research and development programs through our wholly-owned subsidiaries and VIEs that we may ultimately determine not to advance, based on our ongoing assessment of the likelihood of success relative to the costs and risks associated with the program.   

Risks Related to the Clinical Development of Our Product Candidates

We may encounter substantial delays in clinical trials, or may not be able to conduct or complete clinical trials on the expected timelines, if at all.

Clinical testing is expensive, time consuming and subject to uncertainty. We cannot guarantee that any of our ongoing and planned clinical trials will be conducted as planned or completed on schedule, if at all. Moreover, even if these trials are initiated or conducted on a timely basis, issues may arise that could result in the suspension or

53


termination of such clinical trials. A failure of one or more clinical trials can occur at any stage of testing, and our ongoing and future clinical trials may not be successful. Events that may prevent successful or timely initiation or completion of clinical trials include:

 

delays in clinical trial enrollment or clinical trial initiation resulting from the COVID-19 pandemic or any future pandemics;

 

inability to generate sufficient preclinical, toxicology or other in vivo or in vitro data to support the initiation or continuation of clinical trials;

 

delays in confirming target engagement, patient selection or other relevant biomarkers to be utilized in preclinical and clinical product candidate development;

 

delays in reaching a consensus with regulatory agencies as to the design or implementation of our clinical trials;

 

delays in reaching agreement on acceptable terms with prospective CROs and clinical trial sites, the terms of which can be subject to extensive negotiation and may vary significantly among different CROs and clinical trial sites;

 

delays in identifying, recruiting and training suitable clinical investigators;

 

delays in obtaining required Institutional Review Board, or IRB, approval at each clinical trial site;

 

imposition of a temporary or permanent clinical hold by regulatory agencies for a number of reasons, including after review of an Investigational New Drug application, or IND, or IND amendment, clinical trial application, or CTA, or CTA amendment, or equivalent application or amendment; or as a result of a new safety finding that presents unreasonable risk to clinical trial participants or a negative finding from an inspection of our clinical trial operations or study sites;

 

developments in trials for other product candidates with the same targets or related modalities as our product candidates conducted by competitors that raise regulatory or safety concerns about risk to patients of the treatment, or if the FDA finds that the investigational protocol or plan is clearly deficient to meet its stated objectives;

 

difficulties in securing access to materials for the comparator arm of certain of our clinical trials;

 

delays in identifying, recruiting and enrolling suitable patients to participate in clinical trials, and delays caused by patients withdrawing from clinical trials or failing to return for post-treatment follow-up;

 

difficulty collaborating with patient groups and investigators;

 

failure by CROs, other third parties or us to adhere to clinical trial requirements;

 

failure to perform in accordance with the FDA’s or any other regulatory authority’s current good clinical practices, or GCP, requirements, or regulatory guidelines in other countries;

 

occurrence of adverse events, or AEs, associated with the product candidate that are viewed to outweigh its potential benefits;

 

changes in regulatory requirements and guidance that require amending or submitting new clinical protocols;

 

changes in the standard of care on which a clinical development plan was based, which may require new or additional trials;