false2021Q20001583648December 310000000000015836482021-01-012021-06-30xbrli:shares00015836482021-08-02iso4217:USD00015836482021-06-3000015836482020-12-3100015836482021-04-012021-06-3000015836482020-04-012020-06-3000015836482020-01-012020-06-30iso4217:USDxbrli:shares0001583648us-gaap:PreferredStockMember2020-03-310001583648us-gaap:CommonStockMember2020-03-310001583648us-gaap:AdditionalPaidInCapitalMember2020-03-310001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-03-310001583648us-gaap:RetainedEarningsMember2020-03-3100015836482020-03-310001583648us-gaap:RetainedEarningsMember2020-04-012020-06-300001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-04-012020-06-300001583648us-gaap:AdditionalPaidInCapitalMember2020-04-012020-06-300001583648us-gaap:CommonStockMember2020-04-012020-06-300001583648us-gaap:SeriesDPreferredStockMemberus-gaap:PreferredStockMember2020-04-012020-06-300001583648us-gaap:CommonStockMemberus-gaap:SeriesDPreferredStockMember2020-04-012020-06-300001583648us-gaap:SeriesDPreferredStockMemberus-gaap:AdditionalPaidInCapitalMember2020-04-012020-06-300001583648us-gaap:SeriesDPreferredStockMember2020-04-012020-06-300001583648us-gaap:PreferredStockMember2020-06-300001583648us-gaap:CommonStockMember2020-06-300001583648us-gaap:AdditionalPaidInCapitalMember2020-06-300001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-06-300001583648us-gaap:RetainedEarningsMember2020-06-3000015836482020-06-300001583648us-gaap:PreferredStockMember2021-03-310001583648us-gaap:CommonStockMember2021-03-310001583648us-gaap:AdditionalPaidInCapitalMember2021-03-310001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-03-310001583648us-gaap:RetainedEarningsMember2021-03-3100015836482021-03-310001583648us-gaap:RetainedEarningsMember2021-04-012021-06-300001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-04-012021-06-300001583648us-gaap:AdditionalPaidInCapitalMember2021-04-012021-06-300001583648us-gaap:CommonStockMember2021-04-012021-06-300001583648us-gaap:PreferredStockMember2021-04-012021-06-300001583648us-gaap:PreferredStockMemberus-gaap:SeriesEPreferredStockMember2021-04-012021-06-300001583648us-gaap:CommonStockMemberus-gaap:SeriesEPreferredStockMember2021-04-012021-06-300001583648us-gaap:AdditionalPaidInCapitalMemberus-gaap:SeriesEPreferredStockMember2021-04-012021-06-300001583648us-gaap:SeriesEPreferredStockMember2021-04-012021-06-300001583648pirs:AtTheMarketOfferingMember2021-04-012021-06-300001583648pirs:AtTheMarketOfferingMemberus-gaap:CommonStockMember2021-04-012021-06-300001583648pirs:AtTheMarketOfferingMemberus-gaap:AdditionalPaidInCapitalMember2021-04-012021-06-300001583648us-gaap:PrivatePlacementMember2021-04-012021-06-300001583648us-gaap:CommonStockMemberus-gaap:PrivatePlacementMember2021-04-012021-06-300001583648us-gaap:PrivatePlacementMemberus-gaap:AdditionalPaidInCapitalMember2021-04-012021-06-300001583648us-gaap:PreferredStockMember2021-06-300001583648us-gaap:CommonStockMember2021-06-300001583648us-gaap:AdditionalPaidInCapitalMember2021-06-300001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-06-300001583648us-gaap:RetainedEarningsMember2021-06-300001583648us-gaap:PreferredStockMember2019-12-310001583648us-gaap:CommonStockMember2019-12-310001583648us-gaap:AdditionalPaidInCapitalMember2019-12-310001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2019-12-310001583648us-gaap:RetainedEarningsMember2019-12-3100015836482019-12-310001583648us-gaap:RetainedEarningsMember2020-01-012020-06-300001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-01-012020-06-300001583648us-gaap:AdditionalPaidInCapitalMember2020-01-012020-06-300001583648us-gaap:CommonStockMember2020-01-012020-06-300001583648us-gaap:SeriesDPreferredStockMemberus-gaap:PreferredStockMember2020-01-012020-06-300001583648us-gaap:CommonStockMemberus-gaap:SeriesDPreferredStockMember2020-01-012020-06-300001583648us-gaap:SeriesDPreferredStockMemberus-gaap:AdditionalPaidInCapitalMember2020-01-012020-06-300001583648us-gaap:SeriesDPreferredStockMember2020-01-012020-06-300001583648us-gaap:PreferredStockMember2020-12-310001583648us-gaap:CommonStockMember2020-12-310001583648us-gaap:AdditionalPaidInCapitalMember2020-12-310001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2020-12-310001583648us-gaap:RetainedEarningsMember2020-12-310001583648us-gaap:RetainedEarningsMember2021-01-012021-06-300001583648us-gaap:AccumulatedOtherComprehensiveIncomeMember2021-01-012021-06-300001583648us-gaap:AdditionalPaidInCapitalMember2021-01-012021-06-300001583648us-gaap:CommonStockMember2021-01-012021-06-300001583648us-gaap:PreferredStockMember2021-01-012021-06-300001583648us-gaap:PreferredStockMemberus-gaap:SeriesEPreferredStockMember2021-01-012021-06-300001583648us-gaap:CommonStockMemberus-gaap:SeriesEPreferredStockMember2021-01-012021-06-300001583648us-gaap:AdditionalPaidInCapitalMemberus-gaap:SeriesEPreferredStockMember2021-01-012021-06-300001583648us-gaap:SeriesEPreferredStockMember2021-01-012021-06-300001583648pirs:AtTheMarketOfferingMember2021-01-012021-06-300001583648pirs:AtTheMarketOfferingMemberus-gaap:CommonStockMember2021-01-012021-06-300001583648pirs:AtTheMarketOfferingMemberus-gaap:AdditionalPaidInCapitalMember2021-01-012021-06-300001583648us-gaap:PrivatePlacementMember2021-01-012021-06-300001583648us-gaap:CommonStockMemberus-gaap:PrivatePlacementMember2021-01-012021-06-300001583648us-gaap:PrivatePlacementMemberus-gaap:AdditionalPaidInCapitalMember2021-01-012021-06-30xbrli:pure0001583648pirs:PierisPharmaceuticalsGmbHMember2013-05-310001583648srt:MinimumMemberpirs:LaboratoryEquipmentMember2021-01-012021-06-300001583648pirs:LaboratoryEquipmentMembersrt:MaximumMember2021-01-012021-06-300001583648srt:MinimumMemberus-gaap:OfficeEquipmentMember2021-01-012021-06-300001583648us-gaap:OfficeEquipmentMembersrt:MaximumMember2021-01-012021-06-300001583648srt:MinimumMemberus-gaap:ComputerEquipmentMember2021-01-012021-06-300001583648us-gaap:ComputerEquipmentMembersrt:MaximumMember2021-01-012021-06-300001583648pirs:SeattleGeneticsInc.Member2021-04-012021-06-300001583648pirs:SeattleGeneticsInc.Member2020-04-012020-06-300001583648pirs:SeattleGeneticsInc.Member2021-01-012021-06-300001583648pirs:SeattleGeneticsInc.Member2020-01-012020-06-300001583648pirs:AstraZenecaABMember2021-04-012021-06-300001583648pirs:AstraZenecaABMember2020-04-012020-06-300001583648pirs:AstraZenecaABMember2021-01-012021-06-300001583648pirs:AstraZenecaABMember2020-01-012020-06-300001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2021-04-012021-06-300001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2020-04-012020-06-300001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2021-01-012021-06-300001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2020-01-012020-06-300001583648pirs:AstraZenecaABMemberus-gaap:CollaborativeArrangementMemberpirs:ResearchAndDevelopmentMilestonePaymentsMember2021-06-300001583648pirs:AstraZenecaABMemberus-gaap:CollaborativeArrangementMemberpirs:SalesMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMemberpirs:ResearchAndDevelopmentMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:SalesMilestonePaymentsMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2021-06-300001583648pirs:SeattleGeneticsInc.Memberus-gaap:CollaborativeArrangementMemberpirs:ResearchAndDevelopmentMilestonePaymentsMember2021-06-300001583648pirs:SeattleGeneticsInc.Memberus-gaap:CollaborativeArrangementMemberpirs:SalesMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:BostonPharmaceuticalsMemberpirs:ResearchAndDevelopmentMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:BostonPharmaceuticalsMemberpirs:SalesMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:GenentechMemberpirs:ResearchAndDevelopmentMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:GenentechMemberpirs:SalesMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:ResearchAndDevelopmentMilestonePaymentsMember2021-06-300001583648us-gaap:CollaborativeArrangementMemberpirs:SalesMilestonePaymentsMember2021-06-300001583648us-gaap:LicenseMemberpirs:GenentechMember2021-05-190001583648pirs:GenentechMember2021-05-190001583648pirs:GenentechMember2021-05-192021-05-19pirs:leaseAgreement0001583648pirs:GenentechMemberpirs:ResearchAndDevelopmentServicesMember2021-05-192021-05-19pirs:performanceObligation0001583648pirs:LicenseAndCollaborationAgreementMemberpirs:GenentechMember2021-05-192021-05-190001583648pirs:GenentechMemberpirs:PlatformTechnologyLicenseMember2021-05-192021-05-19pirs:program0001583648pirs:GenentechMemberpirs:AntibodyTargetSwapMember2021-05-192021-05-190001583648pirs:GenentechMemberpirs:GovernanceCommitteeParticipationMember2021-05-192021-05-190001583648pirs:GenentechMember2021-06-300001583648us-gaap:LicenseMemberpirs:GenentechMember2021-06-300001583648us-gaap:LicenseMemberpirs:ExclusiveProductLicenseAgreementMemberpirs:BPAssetsXIIIncMember2021-04-240001583648us-gaap:LicenseMemberpirs:ExclusiveProductLicenseAgreementMemberpirs:BPAssetsXIIIncMember2021-04-242021-04-240001583648us-gaap:LicenseMemberpirs:BPAssetsXIIIncMemberus-gaap:UpFrontPaymentArrangementMember2021-04-240001583648us-gaap:LicenseMemberpirs:BPAssetsXIIIncMember2021-04-240001583648us-gaap:LicenseMemberpirs:ExclusiveProductLicenseAgreementMemberpirs:BPAssetsXIIIncMember2021-06-300001583648us-gaap:LicenseMember2021-06-30pirs:researchProgram0001583648pirs:SeattleGeneticsInc.Memberpirs:LicenseAndCollaborationAgreementMember2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Memberpirs:OtherArrangementMember2018-02-082018-02-080001583648us-gaap:LicenseMemberpirs:SeattleGeneticsInc.Memberus-gaap:UpFrontPaymentArrangementMember2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Memberus-gaap:UpFrontPaymentArrangementMember2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Memberus-gaap:UpFrontPaymentArrangementMember2018-02-08pirs:licensepirs:swapOption0001583648pirs:SeattleGeneticsInc.Memberpirs:PlatformTechnologyLicenseMember2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Memberpirs:AntibodyTargetSwapMember2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Memberpirs:GovernanceCommitteeParticipationMember2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Member2018-02-082018-02-080001583648pirs:SeattleGeneticsInc.Member2021-06-300001583648pirs:SeattleGeneticsInc.Memberpirs:SalesMilestonePaymentsMemberus-gaap:UpFrontPaymentArrangementMember2020-01-012020-12-310001583648pirs:SeattleGeneticsInc.Memberpirs:SalesMilestonePaymentsMemberus-gaap:UpFrontPaymentArrangementMember2020-04-012020-06-300001583648pirs:SeattleGeneticsInc.Memberus-gaap:PrivatePlacementMember2021-03-242021-03-240001583648pirs:SeattleGeneticsInc.Memberus-gaap:PrivatePlacementMember2021-03-240001583648pirs:SeattleGeneticsInc.Member2021-03-242021-03-24pirs:protein0001583648pirs:AstraZenecaABMemberpirs:LicenseAndCollaborationAgreementMember2017-05-022017-05-02pirs:collaborationProduct0001583648srt:MinimumMemberpirs:AstraZenecaABMemberpirs:LicenseAndCollaborationAgreementMember2017-05-020001583648pirs:AstraZenecaABMemberpirs:LicenseAndCollaborationAgreementMembersrt:MaximumMember2017-05-020001583648pirs:AstraZenecaABMemberpirs:LicenseAndCollaborationAgreementMembersrt:MaximumMember2017-05-022017-05-020001583648pirs:AstraZenecaABMemberus-gaap:UpFrontPaymentArrangementMember2017-05-022017-05-0200015836482017-05-022017-05-020001583648pirs:AstraZenecaABMemberpirs:EstimatedDevelopmentAndManufacturingServicesMemberus-gaap:UpFrontPaymentArrangementMember2017-05-022017-05-020001583648pirs:AstraZenecaABMemberpirs:SalesMilestonePaymentsMemberus-gaap:UpFrontPaymentArrangementMember2017-05-022017-05-020001583648pirs:AstraZenecaABMemberpirs:EstimatedPhase2aServicesMemberus-gaap:UpFrontPaymentArrangementMember2017-05-022017-05-020001583648pirs:AstraZenecaABMemberpirs:AdditionalOtherResearchServicesMember2017-05-022017-05-020001583648pirs:AstraZenecaABMember2017-05-022017-05-020001583648pirs:AstraZenecaABMember2021-06-300001583648pirs:AstraZenecaABMemberus-gaap:PrivatePlacementMember2021-04-012021-04-010001583648pirs:AstraZenecaABMemberus-gaap:PrivatePlacementMember2021-04-010001583648pirs:AstraZenecaABMember2021-04-012021-04-010001583648pirs:AstraZenecaABMember2021-03-012021-03-310001583648pirs:AstraZenecaABMember2021-01-012021-03-310001583648pirs:AstraZenecaABMemberus-gaap:AccountingStandardsUpdate201409Member2017-05-020001583648pirs:AstraZenecaABMemberus-gaap:AccountingStandardsUpdate201409Member2019-01-010001583648pirs:AstraZenecaABMemberus-gaap:AccountingStandardsUpdate201409Member2021-06-30pirs:bispecificTherapeuticProgram0001583648pirs:LicenseAndCollaborationAgreementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-040001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-04iso4217:EUR0001583648us-gaap:LicenseMemberus-gaap:UpFrontPaymentArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-04pirs:preclinicalMilestone0001583648pirs:ResearchAndDevelopmentServicesMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-040001583648pirs:ResearchAndDevelopmentServicesMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2018-12-012018-12-310001583648us-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2018-12-012018-12-310001583648us-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2019-02-012019-02-28pirs:extensionOption0001583648pirs:ResearchAndDevelopmentServicesMemberpirs:ExtensionTermMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-040001583648us-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-040001583648srt:MinimumMemberus-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-040001583648srt:MaximumMemberus-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-042017-01-040001583648pirs:AstraZenecaABMemberpirs:LicenseAndCollaborationAgreementMembersrt:MaximumMember2017-01-042017-01-040001583648pirs:AstraZenecaABMemberpirs:LicenseAndCollaborationAgreementMembersrt:MaximumMember2017-01-0400015836482017-01-042017-01-04pirs:product0001583648us-gaap:CollaborativeArrangementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-040001583648pirs:LicenseAndCollaborationAgreementMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2017-01-040001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2021-06-300001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2020-02-012020-02-290001583648pirs:CoDevelopmentCollaborationProductMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2020-02-290001583648pirs:ServierDevelopedCollaborationProductMemberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2020-02-290001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2020-01-012020-03-310001583648pirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2021-02-012021-02-280001583648us-gaap:AccountingStandardsUpdate201409Memberpirs:LesLaboratoiresServierAndInstitutDeRecherchesInternationalesServierMember2019-01-010001583648pirs:LaboratoryEquipmentMember2021-06-300001583648pirs:LaboratoryEquipmentMember2020-12-310001583648us-gaap:OfficeEquipmentMember2021-06-300001583648us-gaap:OfficeEquipmentMember2020-12-310001583648us-gaap:ComputerEquipmentMember2021-06-300001583648us-gaap:ComputerEquipmentMember2020-12-310001583648us-gaap:LeaseholdImprovementsMember2021-06-300001583648us-gaap:LeaseholdImprovementsMember2020-12-310001583648us-gaap:SeriesAPreferredStockMember2021-06-300001583648us-gaap:SeriesAPreferredStockMember2020-12-310001583648us-gaap:SeriesBPreferredStockMember2021-06-300001583648us-gaap:SeriesBPreferredStockMember2020-12-310001583648us-gaap:SeriesCPreferredStockMember2021-06-300001583648us-gaap:SeriesCPreferredStockMember2020-12-310001583648us-gaap:SeriesDPreferredStockMember2021-06-300001583648us-gaap:SeriesDPreferredStockMember2020-12-310001583648us-gaap:SeriesEPreferredStockMember2021-06-300001583648us-gaap:SeriesEPreferredStockMember2020-12-310001583648pirs:EmployeeDirectorAndConsultantEquityIncentivePlan2020Member2021-06-300001583648pirs:EmployeeDirectorAndConsultantEquityIncentivePlan2019Member2021-01-012021-06-300001583648pirs:EmployeeDirectorAndConsultantEquityIncentivePlan2020Member2021-06-252021-06-250001583648us-gaap:CommonStockMemberpirs:BiotechnologyValueFundLPMember2020-04-012020-04-010001583648us-gaap:SeriesDPreferredStockMemberus-gaap:PreferredStockMember2020-04-012020-04-010001583648us-gaap:SeriesDPreferredStockMemberus-gaap:PreferredStockMember2020-04-010001583648pirs:StockExchangeSharesfromExistingShareholdersMemberus-gaap:CommonStockMember2021-05-202021-05-200001583648pirs:PreferredShareExchangeMemberus-gaap:PreferredStockMemberus-gaap:SeriesEPreferredStockMember2021-05-202021-05-200001583648us-gaap:PreferredStockMemberus-gaap:SeriesEPreferredStockMember2021-05-200001583648pirs:StockExchangeSharesfromExistingShareholdersMemberus-gaap:PreferredStockMemberus-gaap:SeriesEPreferredStockMember2020-04-300001583648pirs:StockExchangeSharesfromExistingShareholdersMemberus-gaap:SeriesCPreferredStockMemberus-gaap:PreferredStockMember2019-11-300001583648pirs:StockExchangeSharesfromExistingShareholdersMemberus-gaap:SeriesBPreferredStockMemberus-gaap:PreferredStockMember2019-01-310001583648pirs:StockExchangeSharesfromExistingShareholdersMemberus-gaap:SeriesAPreferredStockMemberus-gaap:PreferredStockMember2016-06-300001583648us-gaap:PreferredStockMemberus-gaap:SeriesEPreferredStockMember2021-05-202021-05-200001583648pirs:AtTheMarketOfferingMember2019-08-012019-08-310001583648pirs:AtTheMarketOfferingMember2019-08-012021-06-300001583648pirs:AtTheMarketOfferingMember2021-06-30utr:sqft0001583648pirs:BostonMassachusettsMember2015-08-012015-08-310001583648us-gaap:SubsequentEventMemberpirs:BostonMassachusettsMember2021-07-310001583648pirs:FreisingMember2020-01-012020-03-310001583648pirs:LeasesinSameBuildingMemberpirs:FreisingMember2020-01-012020-03-31pirs:floor0001583648pirs:HallbergmoosGermanyMember2020-02-012020-02-290001583648pirs:HallbergmoosGermanyMemberpirs:LeasesExpectedtobeDeliveredbyFebruary2020Member2020-02-012020-02-290001583648pirs:LeasesExpectedtobeDeliveredbyMay2020Memberpirs:HallbergmoosGermanyMember2020-05-012020-05-310001583648pirs:LeasesExpectedtobeDeliveredbyOctober2024Memberpirs:HallbergmoosGermanyMember2021-01-012021-06-300001583648pirs:HallbergmoosGermanyMemberpirs:LeaseswithFirstRightofRefusalforAdditionalAreaMember2021-01-012021-06-300001583648pirs:HallbergmoosGermanyMember2020-02-290001583648pirs:HallbergmoosGermanyMember2021-01-012021-06-300001583648pirs:HallbergmoosGermanyMember2021-06-300001583648pirs:HallbergmoosGermanyMember2018-10-310001583648pirs:HallbergmoosGermanyMember2019-03-31

UNITED STATES
SECURITIES AND EXCHANGE COMMISSION
Washington, D.C. 20549
FORM 10-Q
(Mark One)
QUARTERLY REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934
For the quarterly period ended June 30, 2021
OR
TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934
For the transition period from                      to                     
Commission File Number: 001-37471
PIERIS PHARMACEUTICALS, INC.
(Exact name of registrant as specified in its charter)
Nevada 30-0784346
(State or other jurisdiction of
incorporation or organization)
 (I.R.S. Employer
Identification No.)
255 State Street9th Floor 
Boston,MA
United States02109
(Address of principal executive offices) (Zip Code)
857-246-8998
Registrant’s telephone number, including area code
Securities registered pursuant to Section 12(b) of the Act:
Title of Each ClassTrading SymbolName of Each Exchange on Which Registered
Common Stock, $0.001 par value per sharePIRS
The Nasdaq Capital Market
Indicate by check mark whether the registrant (1) has filed all reports required to be filed by Section 13 or 15(d) of the Securities Exchange Act of 1934 during the preceding 12 months (or for such shorter period that the registrant was required to file such reports), and (2) has been subject to such filing requirements for the past 90 days.    Yes  No 
Indicate by check mark whether the registrant has submitted electronically every Interactive Data File required to be submitted pursuant to Rule 405 of Regulation S-T (§232.405 of this chapter) during the preceding 12 months (or for such shorter period that the registrant was required to submit such files).    Yes  No 
Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, a smaller reporting company or an emerging growth company. See the definitions of “large accelerated filer,” “accelerated filer,” “smaller reporting company” and “emerging growth company” in Rule 12b-2 of the Exchange Act.
Large accelerated filer   Accelerated filer 
Non-accelerated filer 
  Smaller reporting company 
Emerging growth company
1


If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act.
Indicate by check mark whether the registrant is a shell company (as defined in Rule 12b-2 of the Exchange Act).    Yes    No 

As of August 2, 2021, the registrant had 66,710,712 shares of common stock outstanding.
2


TABLE OF CONTENTS
 Page


i


Special Note Regarding Forward-Looking Statements

This Quarterly Report on Form 10-Q contains forward looking statements within the meaning of Section 27A of the Securities Act of 1933, as amended, or the Securities Act, and Section 21E of the Securities Exchange Act of 1934, as amended, or the Exchange Act, that involve risks and uncertainties, principally in the section titled “Management’s Discussion and Analysis of Financial Condition and Results of Operations.” All statements other than statements of historical fact contained in this Quarterly Report on Form 10-Q, including statements regarding future events, our future financial performance, expectations for growth and revenues, anticipated timing and amounts of milestone and other payments under collaboration agreements, business strategy and plans, objectives of management for future operations, timing and outcome of legal and other proceedings, and our ability to finance our operations are forward-looking statements. We have attempted to identify forward-looking statements by terminology including “anticipates,” “believes,” “can,” “continue,” “look forward,” “ongoing,” “could,” “estimates,” “expects,” “intends,” “may,” “appears,” “suggests,” “future,” “likely,” “plans,” “potential,” “possibly,” “projects,” “predicts,” “seek,” “should,” “target,” “would” or “will” or the negative of these terms or other comparable terminology. Although we do not make forward-looking statements unless we believe we have a reasonable basis for doing so, we cannot guarantee their accuracy. These statements are only predictions and involve known and unknown risks, uncertainties and other factors, including the risks outlined under “Risk Factors” or elsewhere in our most recent Annual Report on Form 10-K or Quarterly Reports on Form 10-Q, which may cause our or our industry’s actual results, levels of activity, performance or achievements expressed or implied by these forward-looking statements to differ materially.

Moreover, we operate in a very competitive and rapidly changing environment. New risks emerge from time to time, and it is not possible for us to predict all risk factors, nor can we address the impact of all factors on our business or the extent to which any factor, or combination of factors, may cause our actual results to differ materially from those contained in any forward-looking statements. Actual results could differ materially from our forward-looking statements due to a number of factors, including, without limitation, risks related to: the results of our research and development activities, including uncertainties relating to the discovery of potential drug candidates and the preclinical and ongoing or planned clinical testing of our drug candidates; the early stage of our drug candidates presently under development; our ability to obtain and, if obtained, maintain regulatory approval of our current drug candidates and any of our other future drug candidates; our need for substantial additional funds in order to continue our operations and the uncertainty of whether we will be able to obtain the funding we need; our future financial performance; our ability to retain or hire key scientific or management personnel; our ability to protect our intellectual property rights that are valuable to our business, including patent and other intellectual property rights; our dependence on third-party manufacturers, suppliers, research organizations, testing laboratories and other potential collaborators; the success of our collaborations with third parties; our ability to meet milestones; our ability to successfully market and sell our drug candidates in the future as needed; the size and growth of the potential markets for any of our approved drug candidates and the rate and degree of market acceptance of any of our approved drug candidates; competition in our industry; regulatory developments in the United States and foreign countries, including with respect to the U.S. Food and Drug Administration, or FDA; our ability to advance our phase 2 study for cinrebafusp alfa, or PRS-343; the expected impact of new accounting standards; and the length and severity of the pandemic relating to SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), or coronavirus, which causes coronavirus disease 2019, or COVID-19, which could have an impact on our research, development, supply chain and clinical trials.

You should not place undue reliance on any forward-looking statement(s), each of which applies only as of the date of this Quarterly Report on Form 10-Q. Before you invest in our securities, you should be aware that the occurrence of the events described in Part I, Item 1A (Risk Factors) of our Annual Report on Form 10-K for the fiscal year ended December 31, 2020 filed with the Securities and Exchange Commission, or SEC, on March 31, 2021, could negatively affect our business, operating results, financial condition and stock price. All forward-looking statements included in this document are based on information available to us on the date hereof, and except as required by law, we undertake no obligation to update or revise publicly any of the forward-looking statements after the date of this Quarterly Report on Form 10-Q to conform our statements to actual results or changed expectations.
Currency Presentation and Currency Translation
Unless otherwise indicated, all references to “dollars,” “$,” “U.S. $” or “U.S. dollars” are to the lawful currency of the United States. All references in this Quarterly Report on Form 10-Q to “euro” or “€” are to the currency introduced at the start of the third stage of the European Economic and Monetary Union pursuant to the Treaty establishing the European Community, as amended. We prepare our financial statements in U.S. dollars.
The functional currency for our operations is primarily the euro. With respect to our financial statements, the translation from the euro to U.S. dollars is performed for balance sheet accounts using exchange rates in effect at the balance sheet date and for revenue and expense accounts using a weighted average exchange rate during the period. The resulting translation adjustments are recorded as a component of accumulated other comprehensive income/loss.

Where in this Quarterly Report on Form 10-Q we refer to amounts in euros, we have for your convenience also, in certain cases, provided a conversion of those amounts to U.S. dollars in parentheses. Where the numbers refer to a specific balance sheet account date or financial statement account period, we have used the exchange rate that was used to perform the conversions in connection with the applicable financial statement. In all other instances, unless otherwise indicated, the conversions have been made using the noon buying rate of €1.00 to U.S. $1.1884 based on information provided by Refinitiv as of June 30, 2021.
ii


PART I - FINANCIAL INFORMATION

Item 1.        Financial Statements.
PIERIS PHARMACEUTICALS, INC.
CONDENSED CONSOLIDATED BALANCE SHEETS
(unaudited, in thousands)
 June 30,December 31,
 20212020
Assets
Current assets:
Cash and cash equivalents$119,097 $70,436 
Accounts receivable2,803 1,706 
Prepaid expenses and other current assets5,773 3,579 
Total current assets127,673 75,721 
Property and equipment, net20,373 22,046 
Operating lease right-of-use assets3,861 3,934 
Other non-current assets3,123 3,309 
Total assets$155,030 $105,010 
Liabilities and stockholders’ equity
Current liabilities:
Accounts payable$2,310 $1,787 
Accrued expenses and other current liabilities16,082 7,731 
Deferred revenues, current portion25,536 12,627 
Total current liabilities43,928 22,145 
Deferred revenue, net of current portion49,421 35,900 
Operating lease liabilities14,960 15,932 
Other long-term liabilities 6 
Total liabilities108,309 73,983 
Stockholders’ equity:
Preferred stock  
Common stock67 56 
Additional paid-in capital277,496 242,672 
Accumulated other comprehensive loss234 (295)
Accumulated deficit(231,076)(211,406)
Total stockholders’ equity46,721 31,027 
Total liabilities and stockholders’ equity$155,030 $105,010 
The accompanying notes are an integral part of these condensed consolidated financial statements.

1


PIERIS PHARMACEUTICALS, INC.
CONDENSED CONSOLIDATED STATEMENTS OF OPERATIONS AND COMPREHENSIVE LOSS
(unaudited)
(in thousands, except per share data)
 Three Months Ended June 30,Six Months Ended June 30,
 2021202020212020
Revenue
Customer revenue$2,540 $10,930 $17,406 $19,815 
Collaboration revenue745 316 1,512 4,692 
Total revenue3,285 11,246 18,918 24,507 
Operating expenses
Research and development15,800 11,333 32,362 24,091 
General and administrative4,246 4,568 8,376 8,927 
Total operating expenses20,046 15,901 40,738 33,018 
Loss from operations(16,761)(4,655)(21,820)(8,511)
Other income (expense)
Interest income3 129 6 448 
Grant income796  796  
Other income (expense)464 (424)1,348 (484)
Net loss$(15,498)$(4,950)$(19,670)$(8,547)
Other comprehensive income:
Foreign currency translation40 178 529 830 
Unrealized gain on available-for-sale securities (15) 101 
Comprehensive loss$(15,458)$(4,787)$(19,141)$(7,616)
Net loss per share
Basic and diluted$(0.25)$(0.09)$(0.33)$(0.16)
Weighted average number of common shares outstanding
Basic and diluted61,905 52,371 59,116 53,792 
The accompanying notes are an integral part of these condensed consolidated financial statements.
2


PIERIS PHARMACEUTICALS, INC.
CONDENSED CONSOLIDATED STATEMENTS OF CHANGES IN STOCKHOLDERS’ EQUITY
(unaudited, in thousands)
For the Three Months Ended June 30, 2020 and 2021
 Preferred sharesCommon sharesAdditional
paid-in
capital
Accumulated
other
comprehensive
loss
Accumulated
deficit
Total Stockholders' equity
 No. of
shares
Share
capital
No. of
shares
Share
capital
Balance as of March 31, 202011 $ 55,212 $55 $228,751 $(1,227)$(177,773)$49,806 
Net loss— — — — — — (4,950)(4,950)
Foreign currency translation adjustment— — — — — 178 — 178 
Unrealized gain on investments— — — — — (15)— (15)
Stock based compensation expense— — — — 1,237 — — 1,237 
Issuance of common stock resulting from exercise of stock options— — 139 — 271 — — 271 
Issuance of common stock resulting from purchase of employee stock purchase plan shares— — 47 — 145 — — 145 
Preferred stock conversion (Series D)3 $— (3,000)$(3)$3 $— $— $ 
Balance as of June 30, 202014 $ 52,399 $52 $230,407 $(1,064)$(182,723)$46,672 
Balance as of March 31, 202114 $ 59,719 $60 $253,560 $194 $(215,578)$38,236 
Net loss— — — — — — (15,498)(15,498)
Foreign currency translation adjustment— — — — — 40 — 40 
Stock based compensation expense— — — — 1,326 — — 1,326 
Issuance of common stock resulting from exercise of stock options— — 129 — 272 — — 272 
Issuance of common stock resulting from purchase of employee stock purchase plan shares— — 40 — 96 — — 96 
Issuance of common stock resulting from exercise of warrants— — 1,391 1 836 — — 837 
Issuance of common stock resulting from conversion of preferred stock(4)— 3,812 4 (4)— —  
Preferred stock conversion (Series E)5 — (5,000)(5)5 — —  
Issuance of common stock pursuant to at the market offering program, net of $0.4 million in offering costs
— — 3,004 3 12,169 — — 12,172 
Issuance of common stock pursuant to private placement offering, net of $0.1 million in offering costs
— — 3,584 4 9,236 — — 9,240 
Balance as of June 30, 202116 $ 66,679 $67 $277,496 $234 $(231,076)$46,721 
The accompanying notes are an integral part of these condensed consolidated financial statements.



3


PIERIS PHARMACEUTICALS, INC.
CONDENSED CONSOLIDATED STATEMENTS OF CHANGES IN STOCKHOLDERS’ EQUITY
(unaudited, in thousands)
For the Six Months Ended June 30, 2020 and 2021
 Preferred sharesCommon sharesAdditional
paid-in
capital
Accumulated
other
comprehensive
loss
Accumulated
deficit
Total Stockholders' equity
 No. of
shares
Share
capital
No. of
shares
Share
capital
Balance as of December 31, 201911 $ 55,212 $55 $227,468 $(1,995)$(174,176)$51,352 
Net loss— — — — — — (8,547)(8,547)
Foreign currency translation adjustment— — — — — 830  830 
Unrealized gain on investments— — — — — 101 — 101 
Stock based compensation expense— — — — 2,520  — 2,520 
Issuance of common stock resulting from exercise of stock options— — 139 — 271 — — 271 
Issuance of common stock resulting from purchase of employee stock purchase plan shares— — 47 — 145 — — 145 
Preferred stock conversion (Series D)3 $— (3,000)$(3)$3 $— $— $ 
Balance as of June 30, 202014 $ 52,399 $52 $230,407 $(1,064)$(182,723)$46,672 
Balance as of December 31, 202014 $ 56,003 $56 $242,672 $(295)$(211,406)$31,027 
Net loss— — — — — — (19,670)(19,670)
Foreign currency translation adjustment— — — — — 529 — 529 
Stock based compensation expense— — — — 2,525 — — 2,525 
Issuance of common stock resulting from exercise of stock options— — 139 — 292 — — 292 
Issuance of common stock resulting from purchase of employee stock purchase plan shares— — 40 — 96 — — 96 
Issuance of common stock resulting from exercise of warrants— — 1,391 1 836 — — 837 
Issuance of common stock resulting from conversion of preferred stock(4)— 3,812 4 (4)— —  
Preferred stock conversion (Series E)5 — (5,000)(5)5 — —  
Issuance of common stock pursuant to at the market offering program, net of $0.4 million in offering costs
— — 3,004 3 12,169 — — 12,172 
Issuance of common stock pursuant to private placement offering, net of $0.1 million in offering costs
— — 7,290 8 18,905 — — 18,913 
Balance as of June 30, 202116 $ 66,679 $67 $277,496 $234 $(231,076)$46,721 
The accompanying notes are an integral part of these condensed consolidated financial statements.
4


PIERIS PHARMACEUTICALS, INC.
CONDENSED CONSOLIDATED STATEMENTS OF CASH FLOWS
(unaudited, in thousands)
 Six Months Ended June 30,
 20212020
Operating activities:
Net loss$(19,670)$(8,547)
Adjustments to reconcile net loss to net cash provided by operating activities:
Depreciation and amortization1,242 869 
Right-of-use asset amortization(48)156 
Stock-based compensation2,525 2,520 
Other non-cash transactions(14)343 
Changes in operating assets and liabilities33,733 (20,250)
Net cash provided by (used in) operating activities17,768 (24,909)
Investing activities:
Purchases of property and equipment(257)(2,988)
Proceeds from maturity of investments 47,523 
Purchases of investments (38,525)
Net cash (used in)/provided by investing activities(257)6,010 
Financing activities:
Proceeds from exercise of stock options292 271 
Proceeds from exercise of warrants837  
Proceeds from employee stock purchase plan96 145 
Proceeds from issuance of common stock from private placement, net of issuance costs18,913  
Proceeds from issuance of common stock resulting from ATM sales, net of $0.4M in transaction costs
12,172  
Net cash provided by financing activities32,310 416 
Effect of exchange rate change on cash and cash equivalents(1,160)525 
Net increase (decrease) in cash and cash equivalents48,661 (17,958)
Cash and cash equivalents at beginning of period70,436 62,260 
Cash and cash equivalents at end of period$119,097 $44,302 
Supplemental cash flow disclosures:
Net unrealized gain on investments$ $31 
Property and equipment included in accounts payable$ $329 
The accompanying notes are an integral part of these condensed consolidated financial statements.
5


PIERIS PHARMACEUTICALS, INC.
NOTES TO THE CONDENSED CONSOLIDATED FINANCIAL STATEMENTS
(unaudited)
 
1.     Corporate Information
Pieris Pharmaceuticals, Inc. was founded in May 2013, and acquired 100% interest in Pieris Pharmaceuticals GmbH (formerly Pieris AG, a German company that was founded in 2001) in December 2014. Pieris Pharmaceuticals, Inc. and its wholly-owned subsidiaries, hereinafter collectively Pieris, or the Company, is a clinical-stage biopharmaceutical company that discovers and develops Anticalin-based drugs to target validated disease pathways in unique and transformative ways. Pieris’ corporate headquarters is located in Boston, Massachusetts and its research facility is located in Hallbergmoos, Germany.
Pieris’ clinical pipeline includes an inhaled IL-4Rα antagonist Anticalin protein to treat moderate-to-severe asthma and an immuno-oncology, or IO, bispecific targeting 4-1BB and HER2.
The Company’s core Anticalin technology and platform was developed in Germany, and the Company has partnership arrangements with several major multi-national pharmaceutical companies.
The Company is subject to risks common to companies in the biotechnology industry, including but not limited to, the need for additional capital, risks of failure of preclinical studies and clinical trials, the need to obtain marketing approval and reimbursement for any drug product candidate that it may identify and develop, the need to successfully commercialize and gain market acceptance of its product candidates, dependence on key personnel, protection of proprietary technology, compliance with government regulations, development of technological innovations by competitors, reliance on third-party manufacturers and the ability to transition from pilot-scale production to large-scale manufacturing of products.

As of June 30, 2021, cash and cash equivalents were $119.1 million. The Company’s net loss was $15.5 million and $5.0 million for the quarters ended June 30, 2021 and 2020, respectively. The Company has incurred net losses since inception and had an accumulated deficit of $231.1 million as of June 30, 2021. Net losses and negative cash flows from operations have had, and will continue to have, an adverse effect on the Company’s stockholders’ equity and working capital. The Company expects to continue to incur operating losses for at least the next several years.

The future success of the Company is dependent on its ability to identify and develop its product candidates, expand its corporate infrastructure and ultimately upon its ability to attain profitable operations. The Company has devoted substantially all of its financial resources and efforts to research and development and general and administrative expenses to support such research and development. The Company has several research and development programs underway in varying stages of development, and it expects that these programs will continue to require increasing amounts of cash for development, conducting clinical trials, and testing and manufacturing of product material. Cash necessary to fund operations will increase significantly over the next several years as the Company continues to conduct these activities necessary to pursue governmental regulatory approval of clinical-stage programs and other product candidates.

The Company plans to raise additional capital to fulfill its operating and capital requirements through public or private equity financings, utilization of its current at the market offering program, or ATM Program, strategic collaborations, licensing arrangements and/or the achievement of milestones under its collaborative agreements. The funding requirements of the Company’s operating plans, however, are based on estimates that are subject to risks and uncertainties and may change as a result of many factors currently unknown. Although management continues to pursue these funding plans, there is no assurance that the Company will be successful in obtaining sufficient funding on terms acceptable to the Company to fund continuing operations, if at all. Until such time that the Company can generate substantial product revenues, if ever, the Company expects to finance its cash needs through a combination of equity offerings, debt financings, strategic partnerships, licensing arrangements and government grants. The terms of any future financing may adversely affect the holdings or the rights of the Company’s existing stockholders.

The Company believes that its currently available funds will be sufficient to fund the Company’s operations through at least the next twelve months from the issuance of this Quarterly Report on Form 10-Q. The Company's belief with respect to its ability to fund operations is based on estimates that are subject to risks and uncertainties. If actual results are different from management’s estimates, the Company may need to seek additional funding. If the Company is unable to obtain additional funding on acceptable terms when needed, it may be required to defer or limit some or all of its research, development and/or clinical projects.

6


2.    Summary of Significant Accounting Policies
The Company’s significant accounting policies are described in Note 2—Summary of Significant Accounting Policies, in the Company’s Annual Report on Form 10-K for the fiscal year ended December 31, 2020. There has been one material addition to the significant accounting policies pertaining to the Company's policy on government grant income during the six months ended June 30, 2021.
Unaudited Interim Financial Information
The accompanying unaudited condensed consolidated financial statements included herein have been prepared by the Company in accordance with accounting principles generally accepted in the United States, or U.S. GAAP, for interim financial information and pursuant to the rules and regulations of the SEC. Accordingly, certain information and footnote disclosures normally included in financial statements prepared in accordance with U.S. GAAP have been condensed or omitted pursuant to such rules and regulations. In the opinion of management, all adjustments, consisting of normal recurring adjustments, and disclosures considered necessary for a fair presentation of interim period results have been included. Interim results for the three and six months ended June 30, 2021 are not necessarily indicative of results that may be expected for the year ending December 31, 2021. For further information, refer to the financial statements and footnotes thereto included in the Company’s Annual Report on Form 10-K for the fiscal year ended December 31, 2020, which was filed with the SEC on March 31, 2021.
Basis of Presentation and Use of Estimates
The accompanying condensed consolidated financial statements of Pieris Pharmaceuticals, Inc. and its wholly-owned subsidiaries were prepared in accordance with U.S. GAAP. The condensed consolidated financial statements include the accounts of all subsidiaries. All intercompany balances and transactions have been eliminated.
The preparation of the financial statements in accordance with U.S. GAAP requires management to make estimates, judgments and assumptions that affect the reported amounts of assets, liabilities, revenues, expenses and the related disclosures at the date of the financial statements and during the reporting period. Significant estimates are used for, but are not limited to, revenue recognition; deferred tax assets, deferred tax liabilities and valuation allowances; determination of the incremental borrowing rate to calculate right-of-use assets and lease liabilities; beneficial conversion features; fair value of stock options, preferred stock, and warrants; and various accruals. Management evaluates its estimates on an ongoing basis. Actual results and outcomes could differ materially from management’s estimates, judgments and assumptions.
Cash, Cash Equivalents and Investments
The Company determines the appropriate classification of its investments at the time of purchase. All liquid investments with original maturities of 90 days or less from the purchase date and for which there is an active market are considered to be cash equivalents. The Company’s investments are comprised of money market, asset backed securities, government treasuries and corporate bonds that are classified as available-for-sale in accordance with Financial Accounting Standards Board, or FASB, Accounting Standards Codification, or ASC, 320, Investments—Debt and Equity Securities. The Company classifies investments available to fund current operations as current assets on its balance sheets.

Available-for-sale investments are recorded at fair value, with unrealized gains or losses included in accumulated other comprehensive loss on the Company’s balance sheets. Realized gains and losses are determined using the specific identification method and are included as a component of other income.

The Company reviews investments for other-than-temporary impairment whenever the fair value of an investment is less than the amortized cost and evidence indicates that an investment’s carrying amount is not recoverable within a reasonable period of time. To determine whether an impairment is other-than temporary, the Company considers its intent to sell or whether it is more likely than not that the Company will be required to sell the investment before recovery of the investment’s amortized cost basis. Evidence considered in this assessment includes reasons for the impairment, the severity and the duration of the impairment and changes in value subsequent to period end.
Concentration of Credit Risk and Off-Balance Sheet Risk
The Company has no financial instruments with off-balance sheet risk such as foreign exchange contracts, option contracts or other foreign hedging arrangements. Financial instruments that subject Pieris to concentrations of credit risk include cash and cash equivalents, investments, and accounts receivable. The Company’s cash, cash equivalents, and investments are held in accounts with financial institutions that management believes are creditworthy. The Company’s investment policy includes guidelines on the quality of the institutions and financial instruments and defines allowable investments that the Company believes minimizes the exposure to concentration of credit risk. The Company has not experienced any credit losses in such
7


accounts and does not believe it is exposed to any significant credit risk on these funds. Accounts receivable primarily consist of amounts due under strategic partnership and other license agreements with major multi-national pharmaceutical companies for which the Company does not obtain collateral.
Fair Value Measurement
The Company is required to disclose information on all assets and liabilities reported at fair value that enables an assessment of the inputs used in determining the reported fair values. FASB ASC Topic 820, Fair Value Measurement and Disclosures, established a hierarchy of inputs used in measuring fair value that maximizes the use of observable inputs and minimizes the use of unobservable inputs by requiring that the observable inputs be used when available. Observable inputs are inputs that market participants would use in pricing the financial instrument based on market data obtained from sources independent of the Company. Unobservable inputs are inputs that reflect the Company’s assumptions about the inputs that market participants would use in pricing the financial instrument and are developed based on the best information available in the circumstances. The fair value hierarchy applies only to the valuation inputs used in determining the reported or disclosed fair value of the financial instruments and is not a measure of the investment credit quality. Fair value measurements are classified and disclosed in one of the following three categories:
Level 1 inputs are quoted prices in active markets for identical assets or liabilities that the reporting entity has the ability to access at the measurement date.
Level 2 utilizes quoted market prices in markets that are not active, broker or dealer quotations or alternative pricing sources with reasonable levels of price transparency.
Level 3 inputs are unobservable inputs for the asset or liability in which there is little, if any, market activity for the asset or liability at the measurement date.

To the extent that valuation is based on models or inputs that are less observable or unobservable in the market, the determination of fair value requires more judgment. Accordingly, the degree of judgment exercised by the Company in determining fair value is greatest for instruments categorized in Level 3. A financial instrument’s level within the fair value hierarchy is based on the lowest level of any input that is significant to the fair value measurement.

Financial instruments measured at fair value on a recurring basis include cash equivalents (see Note 4).

An entity may elect to measure many financial instruments and certain other items at fair value at specified election dates. Subsequent unrealized gains and losses on items for which the fair value option has been elected will be reported in net loss. The Company did not elect to measure any additional financial instruments or other items at fair value.
Property and Equipment

Property and equipment are recorded at acquisition cost, less accumulated depreciation and impairment. Depreciation on property and equipment is calculated using the straight-line method over the remaining estimated useful lives of the assets. Maintenance and repairs to these assets are charged to expenses as occurred. The estimated useful life of the different groups of property and equipment is as follows:

Asset Classification  Estimated useful life (in years)
Leasehold improvements  shorter of useful life or remaining life of the lease
Laboratory furniture and equipment  
8 - 14
Office furniture and equipment  
5 - 13
Computer and equipment
3 - 7

Revenue Recognition

The Company has entered into several licensing agreements with collaboration partners for the development of Anticalin therapeutics against a variety of targets. The terms of these agreements provide for the transfer of multiple goods or services which may include: (i) licenses, or options to obtain licenses, to the Company’s Anticalin technology and/or specific programs and (ii) research and development activities to be performed on behalf of or with a collaborative partner. Payments to Pieris under these agreements may include upfront fees (which include license and option fees), payments for research and development activities, payments based upon the achievement of certain milestones and royalties on product sales. There are no
8


performance, cancellation, termination or refund provisions in any of the arrangements that could result in material financial consequences to Pieris.

Collaborative Arrangements

The Company considers the nature and contractual terms of an arrangement and assesses whether the arrangement involves a joint operating activity pursuant to which it is an active participant and exposed to significant risks and rewards with respect to the arrangement. If the Company is an active participant and exposed to the significant risks and rewards with respect to the arrangement, it accounts for these arrangements pursuant to ASC 808, Collaborative Arrangements, or ASC 808, and applies a systematic and rational approach to recognize revenue. The Company classifies payments received as revenue and payments made as a reduction of revenue in the period in which they are earned. Revenue recognized under a collaborative arrangement involving a participant that is not a customer is presented as Collaboration Revenue in the Statement of Operations.

Revenue from Contracts with Customers

In accordance with ASC 606, revenue is recognized when a customer obtains control of promised goods or services. The amount of revenue recognized reflects the consideration to which the Company expects to be entitled in exchange for these goods and services. To achieve this core principle, the Company applies the following five steps: 1) identify the customer contract; 2) identify the contract’s performance obligations; 3) determine the transaction price; 4) allocate the transaction price to the performance obligations; and 5) recognize revenue when or as a performance obligation is satisfied.

The Company evaluates all promised goods and services within a customer contract and determines which of such goods and services are separate performance obligations. This evaluation includes an assessment of whether the good or service is capable of being distinct and whether the good or service is separable from other promises in the contract.  In assessing whether promised goods or services are distinct, the Company considers factors such as the stage of development of the underlying intellectual property and the capabilities of the customer to develop the intellectual property on their own or whether the required expertise is readily available.

Licensing arrangements are analyzed to determine whether the promised goods or services, which often include licenses, research and development services and governance committee services, are distinct or whether they must be accounted for as part of a combined performance obligation. If the license is considered not to be distinct, the license would then be combined with other promised goods or services as a combined performance obligation. If the Company is involved in a governance committee, it assesses whether its involvement constitutes a separate performance obligation. When governance committee services are determined to be separate performance obligations, the Company determines the fair value to be allocated to this promised service.

Certain contracts contain optional and additional items, which are considered marketing offers and are accounted for as separate contracts with the customer if such option is elected by the customer, unless the option provides a material right which would not be provided without entering into the contract.  An option that is considered a material right is accounted for as a separate performance obligation.

The transaction price is determined based on the consideration to which the Company will be entitled in exchange for transferring goods and services to the customer. A contract may contain variable consideration, including potential payments for both milestone and research and development services. For certain potential milestone payments, the Company estimates the amount of variable consideration by using the most likely amount method. In making this assessment, the Company evaluates factors such as the clinical, regulatory, commercial and other risks that must be overcome to achieve the milestone. Each reporting period the Company re-evaluates the probability of achievement of such variable consideration and any related constraints. Pieris will include variable consideration, without constraint, in the transaction price to the extent it is probable that a significant reversal in the amount of cumulative revenue recognized will not occur when the uncertainty associated with the variable consideration is subsequently resolved. For potential research and development service payments, the Company estimates the amount of variable consideration by using the expected value method, including any approved budget updates arising from additional research or development services.

If the contract contains a single performance obligation, the entire transaction price is allocated to the single performance obligation. Contracts that contain multiple performance obligations require an allocation of the transaction price among the performance obligations on a relative standalone selling price basis unless a portion of the transaction price is variable and meets the criteria to be allocated entirely to a performance obligation or to a distinct good or service that forms part of a single performance obligation.

9


The Company allocates the transaction price based on the estimated standalone selling price of the underlying performance obligations or in the case of certain variable consideration to one or more performance obligations. The Company must develop assumptions that require judgment to determine the stand-alone selling price for each performance obligation identified in the contract. The Company utilizes key assumptions to determine the stand-alone selling price, which may include other comparable transactions, pricing considered in negotiating the transaction and the estimated costs to complete the respective performance obligation. Certain variable consideration is allocated specifically to one or more performance obligations in a contract when the terms of the variable consideration relate to the satisfaction of the performance obligation and the resulting amounts allocated to each performance obligation are consistent with the amount the Company would expect to receive for each performance obligation.   

When a performance obligation is satisfied, revenue is recognized for the amount of the transaction price, excluding estimates of variable consideration that are constrained, that is allocated to that performance obligation on a relative standalone selling price basis. Significant management judgment is required in determining the level of effort required under an arrangement and the period over which the Company is expected to complete its performance obligations under an arrangement.

For performance obligations consisting of licenses and other promises, the Company utilizes judgment to assess the nature of the combined performance obligation to determine whether the combined performance obligation is satisfied over time or at a point in time and, if over time, the appropriate method of measuring progress for purposes of recognizing revenue from non- refundable, up-front fees. The Company evaluates the measure of progress each reporting period and, if necessary, adjusts the measure of performance and related revenue recognition. If the license to the Company’s intellectual property is determined to be distinct from the other performance obligations identified in the arrangement, the Company will recognize revenue from non-refundable, up-front fees allocated to the license when the license is transferred to the customer and the customer is able to use and benefit from the license.

Revenue recognized under an arrangement involving a participant that is a customer is presented as Customer Revenue.

Milestones and Royalties

The Company aggregates milestones into four categories: (i) research milestones, (ii) development milestones, (iii) commercial milestones, and (iv) sales milestones. Research milestones are typically achieved upon reaching certain success criteria as defined in each agreement related to developing an Anticalin protein against the specified target. Development milestones are typically reached when a compound reaches a defined phase of clinical research or passes such phase or upon gaining regulatory approvals. Commercial milestones are typically achieved when an approved pharmaceutical product reaches the status for commercial sale, including regulatory approval. Sales milestones are certain defined levels of net sales by the licensee, such as when a product first achieves global sales or annual sales of a specified amount.

There is uncertainty that the events to obtain the research and development milestones will be achieved given the nature of clinical development and the stage of the Company’s technology. The Company has thus determined that all research and development milestones will be constrained until it is deemed probable that a significant revenue reversal will not occur. For revenues from research and development milestones, payments will be recognized consistent with the recognition pattern of the performance obligation to which they relate.

For arrangements that include sales-based royalties, including milestone payments based on the level of sales, and for which the license is deemed to be the predominant item to which the royalties relate, the Company recognizes revenue at the later of (i) when the related sales occur, or (ii) when the performance obligation to which some or all of the royalty has been allocated has been satisfied (or partially satisfied). Commercial milestones and sales royalties are determined by sales or usage-based thresholds and will be accounted for under the royalty recognition constraint as constrained variable consideration.

Contract Balances

The Company recognizes a contract asset when the Company transfers goods or services to a customer before the customer pays consideration or before payment is due, excluding any amounts presented as a receivable (i.e., accounts receivable). A contract asset is an entity’s right to consideration in exchange for goods or services that the entity has transferred to a customer. The contract liabilities (i.e., deferred revenue) primarily relate to contracts where the Company has received payment but has not yet satisfied the related performance obligations.

In the event of an early termination of a collaboration agreement, any contract liabilities would be recognized in the period in which all Company obligations under the agreement have been fulfilled.

10


Costs to Obtain and Fulfill a Contract with a Customer

Certain costs to obtain customer contracts, including success-based fees paid to third-party service providers, and costs to fulfill customer contracts are capitalized in accordance with FASB ASC 340, Other Assets and Deferred Costs, or ASC 340. These costs are amortized to expense on a systemic basis that is consistent with the transfer to the customer of the goods or services to which the asset relates. The Company will expense the amortization of costs to obtain customer contracts to general and administrative expense and costs to fulfill customer contracts to research and development expense.

Government Grants

The Company recognizes grants from governmental agencies when there is reasonable assurance that the Company will comply with the conditions attached to the grant arrangement and the grant will be received. The Company evaluates the conditions of each grant as of each reporting period to evaluate whether the Company has reached reasonable assurance of meeting the conditions of each grant arrangement and that it is expected that the grant will be received as a result of meeting the necessary conditions. Grants are recognized in the consolidated statements of operations on a systematic basis over the periods in which the Company recognizes the related costs for which the government grant is intended to compensate. Specifically, grant income related to research and development costs is recognized as such expenses are incurred. Grant income is included as a separate caption within Other income (expense), net in the consolidated statements of operations.

Leases

In accordance with ASU No. 2016-2, Leases (Topic 842), or ASC 842, and for each of the Company’s leases, the following is recognized: (i) a lease liability, which is a lessee’s obligation to make lease payments arising from a lease, measured on a discounted basis and (ii) a right-of-use asset, which is an asset that represents the lessee’s right to use, or control the use of, a specified asset for the lease term for all leases (with the exception of short-term leases) at the commencement date.

The Company determines if an arrangement is a lease at inception. The Company’s contracts are determined to contain a lease within the scope of ASC 842 when all of the following criteria based on the specific circumstances of the arrangement are met: (1) there is an identified asset for which there are no substantive substitution rights; (2) the Company has the right to obtain substantially all of the economic benefits from the identified asset; and (3) the Company has the right to direct the use of the identified asset.

At the commencement date, operating lease liabilities and their corresponding right-of-use assets are recorded based on the present value of future lease payments over the expected lease term. The Company’s lease agreements do not provide an implicit rate. As a result, the Company utilizes an estimated incremental borrowing rate to discount lease payments, which is based on the rate of interest the Company would have to pay to borrow a similar amount on a collateralized basis over a similar term and based on observable market data points. Certain adjustments to the right-of-use asset may be required for items such as initial direct costs paid or lease incentives received. Operating lease cost is recognized over the expected term on a straight-line basis.

The Company typically only includes an initial lease term in its assessment of a lease agreement. Options to renew a lease are not included in the Company’s assessment unless there is reasonable certainty that the Company will renew. The expected lease term includes noncancellable lease periods and, when applicable, periods covered by an option to extend the lease if the Company is reasonably certain to exercise that option, as well as periods covered by an option to terminate the lease if the Company is reasonably certain not to exercise that option.

Assumptions made by the Company at the commencement date are re-evaluated upon occurrence of certain events, including a lease modification. A lease modification results in a separate contract when the modification grants the lessee an additional right of use not included in the original lease and when lease payments increase commensurate with the standalone price for the additional right of use. When a lease modification results in a separate contract, it is accounted for in the same manner as a new lease.
Recent Accounting Pronouncement Adopted
In December 2019, the FASB issued ASU No. 2019-12, Income Taxes (Topic 740): Simplifying the Accounting for Income Taxes which amends and aims to simplify accounting disclosure requirements regarding a number of topics including intraperiod tax allocation, accounting for deferred taxes when there are changes in consolidation of certain investments, tax basis step up in an acquisition and the application of effective rate changes during interim periods, among other improvements.
11


This standard is effective for fiscal years beginning after December 15, 2020 and was adopted by the Company on January 1, 2021. Adoption of this new standard did not have a material impact on the Company.
Recent Accounting Pronouncements Not Yet Adopted

In June 2016, the FASB issued ASU No. 2016-13, Financial Instruments - Credit Losses (Topic 326): Measurement of Credit Losses on Financial Statements, or ASU 2016-13. ASU 2016-13 significantly changes the impairment model for most financial assets and certain other instruments. The new standard requires that expected credit losses relating to financial assets measured on an amortized cost basis and available-for-sale debt securities be recorded through an allowance for credit losses. It also limits the amount of credit losses to be recognized for available-for-sale debt securities to the amount by which carrying value exceeds fair value, and requires the reversal of previously recognized credit losses if fair value increases. The allowance for credit losses is a valuation account that is deducted from the amortized cost basis of the financial asset(s) to present the net carrying value at the amount expected to be collected on the financial asset.
Subsequently, in November 2018, the FASB issued ASU No. 2018-19, Codification Improvements to Topic 326, Financial Instruments-Credit Losses, which clarifies codification and corrects unintended application of the guidance. In November 2019, the FASB issued ASU No. 2019-11, Codification Improvements to Topic 326, Financial Instruments-Credit Losses, which clarifies or addresses specific issues about certain aspects of ASU 2016-13. In November 2019 the FASB also issued ASU No. 2019-10, Financial Instruments-Credit Losses (Topic 326), Derivatives and Hedging (Topic 815), and Leases (Topic 842): Effective Dates, which delays the effective date of ASU 2016-13 by three years for certain smaller reporting companies such as the Company. The guidance in ASU 2016-13 is effective for the Company for financial statements issued for fiscal years beginning after December 15, 2022 and interim periods within those fiscal years, with early adoption permitted. The Company is still evaluating the impact of the adoption of this standard.
The Company has considered other recent accounting pronouncements and concluded that they are either not applicable to the business or that the effect is not expected to be material to the unaudited condensed consolidated financial statements as a result of future adoption.
3.    Revenue
General
The Company has not generated revenue from product sales. The Company has generated revenue from contracts with customers and revenue from collaboration agreements, which include upfront payments for licenses or options to obtain licenses, payments for research and development services and milestone payments.
The Company recognized revenue from the following strategic partnerships and other license agreements (in thousands):
 Three Months Ended June 30,Six Months Ended June 30,
 2021202020212020
Seagen$338 $8,512 $704 $8,960 
AstraZeneca2,202 2,334 16,702 5,045 
Servier745 400 1,512 10,502 
Total Revenue$3,285 $11,246 $18,918 $24,507 

Under the Company’s existing strategic partnerships and other license agreements, the Company could receive the following potential milestone payments (in millions):
 Research, Development, Regulatory & Commercial MilestonesSales Milestones
AstraZeneca$1,096 $4,275 
Servier239 214 
Seagen754 450 
Boston Pharmaceuticals88 265 
Genentech834 600 
Total potential milestone payments$3,011 $5,804 
12


Strategic Partnerships
Genentech
On May 19, 2021, the Company and Genentech, Inc., or Genentech, entered into a Research Collaboration and License Agreement, or the Genentech Agreement, to discover, develop and commercialize locally delivered respiratory and ophthalmology therapies that leverage the Company’s proprietary Anticalin technology. Upon signing the Genentech Agreement, Genentech paid the Company a $20 million upfront fee. In addition, the Company may be eligible to receive up to approximately $1.4 billion in additional milestone payments across multiple programs, as well as tiered royalty payments on net sales at percentages ranging from the mid-single to low double-digits, subject to certain standard reductions and offsets.

Under the terms of the Genentech Agreement, the Company will be responsible for discovery and preclinical development of two initial programs. The Company will be responsible for research activities following target nomination through the late-stage research go decision. The parties will then collaborate on drug candidate characterization until the development go decision. After the development go decision, Genentech will be responsible for pursing the preclinical and clinical development of each program, and there after, the commercialization efforts. Each party will be responsible for the costs incurred to perform their respective responsibilities. Genentech has an option to expand the collaboration to encompass two additional programs with the payment of a $10 million fee per additional program. If Genentech exercises its option to start additional programs, payment to the Company of additional fees, milestone payments and royalties would result.

Unless earlier terminated, the term of the Genentech Agreement continues until no royalty or other payment obligations are or will become due under the Genentech Agreement. The Genentech Agreement may be terminated (i) by either party based on insolvency or breach by the other party and such insolvency proceeding is not dismissed or such breach is not cured within 90 days; or (ii) after 9 months from the effective date of the Genentech Agreement, by Genentech as a whole or on a product-by-product and/or country-by-country basis upon 90 days prior written notice before the first commercial sale of a product or upon 180 days prior written notice after the first commercial sale of a product.

While the Genentech Agreement allows for up to four research programs, only two research programs are initially identified in the Genentech Agreement. To reach a total of up to four research programs, the Company has granted Genentech options to nominate an additional two collaboration targets of their choosing, subject to the legal availability of the target to be researched. Genentech will have three years after the effective date to nominate the subsequent targets. The Company has also granted Genentech options to replace any of the collaboration targets identified with another target. However, at no point will there be more than four identified collaboration targets for which there are ongoing research programs.

The arrangement with Genentech provides for the transfer of the following goods or services: (i) exclusive research and commercial license for the collaboration programs, (ii) a non-exclusive platform improvement license, (iii) research and development services, (iv) participation in a governance committee, and (v) replacement target options on the first two programs upon a screening failure which were assessed as material rights.

Management evaluated all of the promised goods or services within the contract and determined which such goods and services were separate performance obligations. The Company determined that the licenses granted, at arrangement inception, should be combined with the research and development services to be provided for the related target programs as they are not capable of being distinct. A third party would not be able to provide the research and development services due to the specific nature of the intellectual property and knowledge required to perform the services, and Genentech could not benefit from the licenses without the corresponding services. The Company determined that the participation in the governance committees was distinct as the services could be performed by an outside party.
As a result, management concluded there were five separate performance obligations at the inception of the Genentech Agreement: (i) two combined performance obligations, each comprised of an exclusive research and commercial license, a non-exclusive platform improvement license, and research and development services for the first two Genentech programs, (ii) two performance obligations each comprised of a material right for a target swap option for the first two Genentech programs, and (iii) one performance obligation comprised of the participation on the governance committee.

The Company allocated consideration to the performance obligations based on the relative proportion of their standalone selling prices. The Company developed standalone selling prices for licenses by applying a risk adjusted, net present value, estimate of future potential cash flows approach, which included the cost of obtaining research and development services at arm’s length from a third-party provider, as well as internal full-time equivalent costs to support these services. The Company developed the standalone selling price for committee participation by using management’s estimate of the anticipated participation hours multiplied by a market rate for comparable participants.

13


The transaction price at inception is comprised of fixed consideration of $20.0 million in upfront fees and was allocated to each of the performance obligations based on the relative proportion of their standalone selling prices. The amounts allocated to the performance obligations for the two research programs will be recognized on a proportional performance basis through the completion of each respective estimated research term of the individual research programs. The amounts allocated to the material right for the target options will be recognized either at the time the material right expires or, if exercised, on a proportional performance basis over the estimated research term for that program along with any remaining deferred revenue associated with the replacement target. The amounts allocated to the participation on the committee will be recognized on a straight-line basis over the anticipated research term for all research programs. As of June 30, 2021, there was $19.5 million of aggregate transaction price allocated to remaining performance obligations.

Under the Genentech Agreement, the Company is eligible to receive various research, development, commercial and sales milestones. There is uncertainty that the events to obtain the research and development milestones will be achieved given the nature of clinical development and the stage of the Company’s technology. The Company has determined that all other research and development milestones will be constrained until it is deemed probable that a significant revenue reversal will not occur.

As of June 30, 2021, there were $5.4 million and $14.1 million of current and non-current deferred revenue, respectively, related to the Genentech Agreement.
Boston Pharmaceuticals

On April 24, 2021, the Company and BP Asset XII, Inc., or Boston Pharmaceuticals, a subsidiary of Boston Pharma Holdings, LLC, entered into an Exclusive Product License Agreement, or the BP Agreement, to develop PRS-342, a 4-1BB/GPC3 preclinical immuno-oncology Anticalin-antibody bispecific fusion protein.

Under the terms of the BP Agreement, Boston Pharmaceuticals exclusively licensed worldwide rights to PRS-342. The Company received an upfront payment of $10.0 million and is further entitled to receive up to $352.5 million in development, regulatory and sales-based milestone payments, tiered royalties up to low double-digits on sales of PRS-342 and a percentage of consideration received by Boston Pharmaceuticals in the event of a sublicense of a program licensed under the BP Agreement or a change of control of Boston Pharmaceuticals. The Company will also contribute up to $4.0 million toward manufacturing activities.

The term of the BP Agreement ends upon the expiration of all of Boston Pharmaceuticals’ payment obligations thereunder. The BP Agreement may be terminated by Boston Pharmaceuticals in its entirety for convenience beginning nine months after its effective date upon 60 days’ notice or, for any program under the BP Agreement which has received marketing approval, upon 120 days’ notice. If any program is terminated by Boston Pharmaceuticals, the Company will have full rights to continue such program. The BP Agreement may also be terminated by Boston Pharmaceuticals or the Company for an uncured material breach by the other party upon 180 days’ notice (60 days in the case of non-payment of undisputed amounts due and payable), subject to extension for an additional 180 days in certain cases and subject, in all cases, to dispute resolution procedures. The Agreement may also be terminated due to the other party’s insolvency. The Company may also terminate the BP Agreement if Boston Pharmaceuticals challenges the validity of any patents licensed under the BP Agreement, subject to certain exceptions.

The Company does not have any obligations to assist in the research and development efforts of Boston Pharmaceuticals under the BP Agreement. However, the Company has an obligation to fund up to $4.0 million in costs, including out-of-pocket costs incurred by Boston Pharmaceuticals, in connection with the manufacture of products under the BP Agreement.
The arrangement with Boston Pharmaceuticals provides for the transfer of the following: (i) exclusive license of PRS-342, (ii) non-exclusive Pieris platform license, (iii) initial know-how, (iv) product cell line license, and (v) materials (as each such term is defined under the BP Agreement).

Management evaluated all of the promised goods or services within the BP Agreement and determined which such goods and services were separate performance obligations. The Company determined that the licenses granted, at arrangement inception, should be combined with the transfer of know-how, materials and the product cell line license. Boston Pharmaceuticals could not benefit from the exclusive and non-exclusive licenses without the corresponding transfer of know-how and materials.
As a result, management concluded there was only one combined performance obligation. The transaction price at inception is comprised of fixed consideration of $10.0 million in upfront fees, offset by $4.0 million in consideration payable to Boston Pharmaceuticals to reimburse them for expected out-of-pocket manufacturing costs, for a total transaction price of $6.0 million. Management has assessed the forms of variable consideration within the BP Agreement and concluded that the payments are either constrained by the royalty recognition constraint or because management has assessed the most likely amount associated with the payments as zero.
14



The amounts allocated to the performance obligations did not meet the criteria to be recognized over time on a proportional performance basis and thus will be recognized at a point in time. The Company determined that the performance obligation will be fully satisfied when all of the deliverables in the combined performance obligation are transferred to Boston Pharmaceuticals as that is the point at which Boston Pharmaceuticals can fully use and benefit from the license to PRS-342. The Company expects all of the deliverables to be transferred to Boston Pharmaceuticals in the third quarter of 2021.

As of June 30, 2021, there was $6.0 million of aggregate transaction price allocated to remaining performance obligations. As of June 30, 2021, there was $6.0 million of current deferred revenue related to the BP Agreement.

Seagen

On February 8, 2018, the Company entered into a license and collaboration agreement, or the Seagen Collaboration Agreement, and a non-exclusive Anticalin platform technology license agreement, or the Seagen Platform License, and together with the Seagen Collaboration Agreement, the Seagen Agreements, with Seagen Inc. (formerly Seattle Genetics, Inc.), or Seagen, pursuant to which the parties will develop multiple targeted bispecific IO treatments for solid tumors and blood cancers.

Under the terms of the Seagen Agreements, the companies will pursue multiple antibody-Anticalin fusion proteins during the research phase. The Seagen Agreements provide Seagen a base option to select up to three programs for further development. Prior to the initiation of a pivotal trial, the Company may opt into global co-development and U.S. commercialization of the second program and share in global costs and profits on an equal basis. Seagen will solely develop, fund and commercialize the other two programs. Seagen may also decide to select additional candidates from the initial research phase for further development in return for the payment to the Company of additional fees, milestone payments and royalties.

The Seagen Platform License grants Seagen a non-exclusive license to certain intellectual property related to the Anticalin platform technology.

Upon signing the Seagen Agreements, Seagen paid the Company a $30.0 million upfront fee and an additional $4.9 million was estimated to be paid for research and development services as reimbursement to the Company through the end of the research term. In addition, the Company may receive tiered royalties on net sales up to the low double-digits and up to $1.2 billion in total success-based research, development, commercial and sales milestones payments across the product candidates, depending on the successful development and commercialization of those candidates. If Seagen exercises its option to select additional candidates from the initial research phase for further development, payment to Pieris of additional fees, milestone payments and royalties would result.

The term of each of the Seagen Agreements ends upon the expiration of all of Seagen’s payment obligations under each such agreement. The Seagen Collaboration Agreement may be terminated by Seagen on a product-by-product basis for convenience beginning 12 months after its effective date upon 90 days’ notice or, for any program where a pivotal study has been initiated, upon 180 days’ notice. Any program may be terminated at Seagen’s option. If any program is terminated by Seagen after a predefined preclinical stage, the Company will have full rights to continue such program. If any program is terminated by Seagen prior to such predefined preclinical stage, the Company will have the right to continue to develop such program, but will be obligated to offer a co-development option to Seagen for such program. The Seagen Collaboration Agreement may also be terminated by Seagen or the Company for an uncured material breach by the other party upon 90 days’ notice, subject to extension for an additional 90 days if the material breach relates to diligence obligations and subject, in all cases, to dispute resolution procedures. The Seagen Collaboration Agreement may also be terminated due to the other party’s insolvency and may in certain instances, including for reasons of safety, be terminated on a product-by-product basis. Each party may also terminate the Seagen Agreements if the other party challenges the validity of any patents licensed under the Seagen Agreements, subject to certain exceptions. The Seagen Platform License will terminate upon termination of the Seagen Collaboration Agreement, whether in its entirety or on a product-by-product basis.

The Company determined that the Seagen Agreements should be combined and evaluated as a single arrangement under ASC 606 as they were executed on the same date. The arrangement with Seagen provides for the transfer of the following goods or services: (i) three candidate research licenses that each consist of a non-exclusive platform technology license, a co-exclusive candidate research license, and research and development services, (ii) research, development and manufacturing services associated with each candidate research license, (iii) participation on various governance committees, and (iv) two antibody target swap options which were assessed as material rights.

Management evaluated all of the promised goods or services within the contract and determined which such goods and services were separate performance obligations. The Company determined that the licenses granted, at arrangement inception, should be
15


combined with the research and development services to be provided for the related antibody target programs as they are not capable of being distinct. A third party would not be able to provide the research and development services due to the specific nature of the intellectual property and knowledge required to perform the services, and Seagen could not benefit from the licenses without the corresponding services. The Company determined that the participation on the various governance committees was distinct as the services could be performed by an outside party.
As a result, management concluded there were six separate performance obligations at the inception of the Seagen Agreements: (i) three combined performance obligations, each comprised of a non-exclusive platform technology license, a co-exclusive candidate research license, and research and development services for the first three approved Seagen antibody target programs, (ii) two performance obligations each comprised of a material right for an antibody target swap option for the first and the second approved Seagen antibody target for no additional consideration, and (iii) one performance obligation comprised of the participation on the various governance committees.

The Company allocated consideration to the performance obligations based on the relative proportion of their standalone selling prices. The Company developed standalone selling prices for licenses by applying a risk adjusted, net present value, estimate of future potential cash flows approach, which included the cost of obtaining research and development services at arm’s length from a third-party provider, as well as internal full-time equivalent costs to support these services. The Company developed the standalone selling price for committee participation by using management’s estimate of the anticipated participation hours multiplied by a market rate for comparable participants.

The transaction price at inception is comprised of fixed consideration of $30.0 million in upfront fees and variable consideration of $4.9 million of estimated research and development services to be reimbursed as research and development occurs through the research term. The $30.0 million upfront fee, which represents the fixed consideration in the transaction price, was allocated to each of the performance obligations based on the relative proportion of their standalone selling prices. The $4.9 million in variable consideration related to the research and development services is allocated specifically to the three target program performance obligations based upon the budgeted services for each program.

The amounts allocated to the performance obligations for the three research programs will be recognized on a proportional performance basis through the completion of each respective estimated research term of the individual research programs. The amounts allocated to the material right for the antibody target swap option will be recognized either at the time the material right expires or, if exercised, on a proportional performance basis over the estimated research term for that program. The amounts allocated to the participation on each of the committees will be recognized on a straight-line basis over the anticipated research term for all research programs. As of June 30, 2021, there was $22.7 million of aggregate transaction price allocated to remaining performance obligations.

In June 2020, Seagen and the Company entered into amendments to the Seagen Agreements, or together, the Amendment. The Amendment extended the deadline for Seagen to nominate a second and third antibody target. As a result of the Amendment, which completed the obligations under the research term for the first antibody target, the Company recorded as revenue $4.2 million, which was previously recorded as deferred revenue, for the year ended December 31, 2020. The Company also recorded $5.0 million of milestone revenue due from Seagen during the quarter ended June 30, 2020, as it was no longer deemed probable that a significant reversal of revenue would occur, and the remaining performance obligations on first antibody target were completed.

On March 24, 2021, the Company announced that Seagen made a strategic equity investment in Pieris, and that the companies had entered into a combination study agreement, or the Combination Study Agreement, to evaluate the safety and efficacy of combining Pieris’ cinrebafusp alfa with Seagen’s tucatinib, a small-molecule tyrosine kinase HER2 inhibitor, for the treatment of gastric cancer patients expressing lower HER2 levels (IHC2+/ISH- & IHC1+) as part of the upcoming phase 2 study to be conducted by Pieris. The companies have also entered into an Amended and Restated License and Collaboration Agreement, or the Second Seagen Amendment, in which their existing IO collaboration agreement has been amended relating to joint development and commercial rights for the second program in the alliance. In connection with the agreements described above, the Company and Seagen also entered into a subscription agreement, or the Seagen Subscription Agreement.

Under the Second Seagen Amendment, Pieris’ option to co-develop and co-commercialize the second of three programs in the collaboration has been converted to a co-promotion option in the United States, with Seagen solely responsible for the development and overall commercialization of that program. Pieris will also be entitled to increased royalties from that program in the event that it chooses to exercise the co-promotion option. In connection with the Seagen Subscription Agreement, the Company agreed to issue to Seagen, and Seagen agreed to acquire from the Company, 3,706,174 shares of the Company’s common stock for a total purchase price of $13.0 million, or $3.51 per share, in a private placement transaction pursuant to Section 4(a)(2) of the Securities Act. The Seagen Subscription Agreement includes a provision to the effect that Seagen may ask the Company to file a registration statement to register the resale of the shares issued to Seagen, at any time beginning on
16


the date that is 60 calendar days from the date of issuance of the shares. The Company assessed the ASC 606 implications of the Seagen Subscription Agreement and concluded that the fair value of the shares on a per share basis was $2.61 per share as of the transaction date. This resulted in a premium paid for the shares of $3.3 million, all of which was recorded in deferred revenue upon contract execution and allocated to the remaining performance obligations.

The Company has concluded that the Combination Study Agreement is within the scope of ASC 808, which defines collaborative arrangements and addresses the presentation of the transactions between the two parties in the income statement and related disclosures. However, ASC 808 does not provide guidance on the recognition of consideration exchanged or accounting for the obligations that may arise between the parties. The Company has concluded that ASC 730, Research and Development, should be applied by analogy. There is no financial statement impact for the Combination Study Agreement as the value of the drug supply received from Seagen is offset against the drug supply cost.

Under the Seagen Agreements, the Company is eligible to receive other various research, development, commercial and sales milestones. There is uncertainty that the events to obtain the research and development milestones will be achieved given the nature of clinical development and the stage of the Company’s technology. With the exception of the previously discussed achieved milestone, the Company has determined that all other research and development milestones will be constrained until it is deemed probable that a significant revenue reversal will not occur.

As of June 30, 2021, there were $6.7 million and $12.8 million of current and non-current deferred revenue, respectively, related to the Seagen Agreements.

AstraZeneca

On May 2, 2017, the Company entered into a license and collaboration agreement, or the AstraZeneca Collaboration Agreement, and a non-exclusive Anticalin platform technology license agreement, or AstraZeneca Platform License, and together with the AstraZeneca Collaboration Agreement, the AstraZeneca Agreements, with AstraZeneca AB, or AstraZeneca, which became effective on June 10, 2017, following expiration of the waiting period under the Hart-Scott-Rodino Antitrust Improvements Act of 1976. Under the AstraZeneca Agreements, the parties will advance several novel inhaled Anticalin proteins.

In addition to the Company’s lead inhaled drug candidate, PRS-060/AZD1402, or the AstraZeneca Lead Product, the Company and AstraZeneca will also collaborate to progress four additional novel Anticalin proteins against undisclosed targets for respiratory diseases, or the AstraZeneca Collaboration Products, and together with the AstraZeneca Lead Product, the AstraZeneca Products. The Company is responsible for advancing the AstraZeneca Lead Product through its phase 1 study, with the associated costs funded by AstraZeneca. The parties will collaborate thereafter to conduct a phase 2a study in asthma patients, with AstraZeneca continuing to fund development costs. After completion of a phase 2a study, Pieris has the option to co-develop the AstraZeneca Lead Product and also has a separate option to co-commercialize the AstraZeneca Lead Product in the United States. For the AstraZeneca Collaboration Products, the Company will be responsible for the initial discovery of the novel Anticalin proteins, after which AstraZeneca will take the lead on continued development of the AstraZeneca Collaboration Products. The Company has the option to co-develop two of the four AstraZeneca Collaboration Products beginning at a predefined preclinical stage and would also have the option to co-commercialize these two programs in the United States, while AstraZeneca will be responsible for development and commercialization of the other programs worldwide.

The term of each of the AstraZeneca Agreements ends upon the expiration of all of AstraZeneca’s payment obligations under such agreement. The AstraZeneca Collaboration Agreement may be terminated by AstraZeneca in its entirety for convenience beginning 12 months after its effective date upon 90 days’ notice or, if the Company has obtained marketing approval for the marketing and sale of a product, upon 180 days’ notice. Each program may be terminated at AstraZeneca’s option; if any program is terminated by AstraZeneca, the Company will have full rights to such program. The AstraZeneca Collaboration Agreement may also be terminated by AstraZeneca or the Company for material breach upon 180 days’ notice of a material breach (or 30 days with respect to payment breach), provided that the applicable party has not cured such breach by the permitted cure period (including an additional 180 days if the breach is not susceptible to cure during the initial 180-day period) and dispute resolution procedures specified in the agreement have been followed. The AstraZeneca Collaboration Agreement may also be terminated due to the other party’s insolvency and may in certain instances be terminated on a product-by-product and/or country-by-country basis. Each party may also terminate an AstraZeneca Agreement if the other party challenges the validity of patents related to certain intellectual property licensed under such AstraZeneca Agreement, subject to certain exceptions for infringement suits, acquisitions and newly-acquired licenses. The AstraZeneca Platform License will terminate upon termination of the AstraZeneca Collaboration Agreement, on a product-by-product and/or country-by-country basis.

17


At inception, AstraZeneca is granted the following licenses: (i) research and development license for the AstraZeneca Lead Product, (ii) commercial license for the AstraZeneca Lead Product, (iii) individual research licenses for each of the four AstraZeneca Collaboration Products, (iv) individual commercial licenses for each of the four AstraZeneca Collaboration Products, and (v) individual non-exclusive platform technology licenses for the AstraZeneca Lead Product and the four AstraZeneca Collaboration Products. AstraZeneca will be granted individual development licenses for each of the four AstraZeneca Collaboration Products upon completion of the initial discovery of Anticalin proteins.

The collaboration will be managed on an overall basis by a Joint Steering Committee, or JSC, formed by an equal number of representatives from the Company and AstraZeneca. In addition to the JSC, the AstraZeneca Collaboration Agreement also requires each party to designate an alliance manager to facilitate communication and coordination of the parties’ activities under the agreement, and further requires participation of both parties on a joint development committee, or JDC, and a commercialization committee. The responsibilities of these committees vary, depending on the stage of development and commercialization of each product.

Under the AstraZeneca Agreements, the Company received an upfront, non-refundable payment of $45.0 million. In addition, the Company will receive payments to conduct a phase 1 clinical study for the AstraZeneca Lead Product. The Company is also eligible to receive research, development, commercial, sales milestone payments and royalty payments. The Company may receive tiered royalties on sales of potential products commercialized by AstraZeneca and for co-developed products, gross margin share on worldwide sales equal to the Company’s level of committed investment.


The Company determined that the AstraZeneca Agreements should be combined and evaluated as a single arrangement under ASC 606 as they were executed on the same date. The arrangement with AstraZeneca, including the impact of any modifications, provides for the transfer of the following goods and services: (i) five non-exclusive platform technology licenses, (ii) research and development license for the AstraZeneca Lead Product, (iii) commercial license for the AstraZeneca Lead Product, (iv) development and manufacturing services for the AstraZeneca Lead Product (or the phase 1 services), (v) technology transfer services for the AstraZeneca Lead Product, (vi) research services related to the AstraZeneca Lead Product, (vii) participation on each of the committees, (viii) four research licenses for the AstraZeneca Collaboration Products, (ix) four commercial licenses for the AstraZeneca Collaboration Products, (x) research services for the AstraZeneca Collaboration Products and (xi) certain phase 2a services for the AstraZeneca Lead Product. Additionally, as the development licenses on the four AstraZeneca Collaboration Products may be granted at a discount in the future, the Company determined such discounts should be assessed as material rights at inception.

Management evaluated all of the promised goods or services within the contract and determined which such goods and services
were separate performance obligations. The Company determined that the licenses granted for the AstraZeneca Lead Product at the inception of the arrangement should be combined with the research services related to the AstraZeneca Lead Product and that the licenses granted for the AstraZeneca Collaboration Products should be combined with the research services for the AstraZeneca Collaboration Products, as the licenses are not capable of being distinct. A third party would not be able to provide the research and development services, due to the specific nature of the intellectual property and knowledge required to perform the services, and AstraZeneca could not benefit from the licenses without the corresponding services. The Company also determined that each of the phase 1 services and the phase 2a services for the AstraZeneca Lead Product were distinct and that the participation on the various committees was also distinct, as all of the phase 1 services, phase 2a services and the committee services could be performed by an outside party. The Company determined that the commercial licenses for the AstraZeneca Collaboration Products granted at the inception of the arrangement should be combined with the development licenses for the AstraZeneca Collaboration Products as the company would not benefit from the commercial license without the ability to develop each product.

As a result, management concluded that there were 16 performance obligations: (i) combined performance obligation comprised of a non-exclusive platform technology license, research and development license, and commercial licenses for the AstraZeneca Lead Product and research services for the AstraZeneca Lead Product, (ii) combined performance obligation comprised of development and manufacturing services, and technology transfer services for the AstraZeneca Lead Product, (iii) committee participation, (iv-vii) four combined performance obligations each comprised of a non-exclusive platform technology license, research licenses, and research services for each AstraZeneca Collaboration Product, (viii-xi) four performance obligations comprised of a material right to acquire the development licenses granted for the AstraZeneca Collaboration Products, (xii-xv) four performance obligations comprised of the commercial licenses granted for the AstraZeneca Collaboration Products and (xvi) phase 2a services.

The Company allocated consideration to the performance obligations based on the relative proportion of their standalone selling prices. The Company developed standalone selling prices for licenses and corresponding research services by applying a risk adjusted, net present value, estimate of future potential cash flow approach, which included the cost of obtaining research
18


services at arm’s length from a third-party provider, as well as internal full-time equivalent costs to support these services. The Company developed its standalone selling price for development and manufacturing services and technology transfer services for the AstraZeneca Lead Product using estimated internal and external costs to be incurred.

The Company developed its standalone selling price for committee participation by using management’s estimate of the anticipated participation hours multiplied by a market rate for comparable participants.

The Company developed its standalone selling price for the commercial licenses and material rights granted on the development licenses by probability weighting multiple cash flow scenarios using the income approach.

The transaction price was comprised of fixed consideration of $45.0 million in upfront fees and variable consideration of (i) $14.2 million in estimated phase 1 services, (ii) $12.5 million in milestone payments achieved upon the initiation of a phase 1 study in December 2017, and (iii) $4.7 million in estimated phase 2a services. The $45.0 million upfront fee, which represents the fixed consideration in the transaction price, was allocated to each of the performance obligations based on the relative proportion of their standalone selling prices. Variable consideration of $14.2 million is related to the phase 1 services and will be allocated entirely to the performance obligation to which they relate. Variable consideration of $12.5 million related to the phase 1 trial milestone was allocated by relative selling price to the combined performance obligation comprised of a non-exclusive platform technology license, research and development license and commercial licenses for the AstraZeneca Lead Product and research services for the AstraZeneca Lead Product, and the combined performance obligation comprised of development and manufacturing services and technology transfer services for the AstraZeneca Lead Product performance obligations. Variable consideration of $4.7 million for phase 2a services was allocated specifically to the related performance obligation.

The amounts allocated to the license performance obligation for the AstraZeneca Lead Product and the four performance obligations for the four research licenses for AstraZeneca Collaboration Products will be recognized on a proportional performance basis as the activities are conducted over the life of the arrangement. The amounts allocated to the performance obligation for phase 1 services, technology transfer services for the AstraZeneca Lead Product will be recognized on a proportional performance basis over the estimated term of development through phase 2a study. The amounts allocated to the performance obligation for phase 2a services for the AstraZeneca Lead Product will be recognized on a proportionate performance basis over an estimated term of 12 months. The amounts allocated to the performance obligation for participation on each of the committees will be recognized on a straight-line basis over the expected term of development of the AstraZeneca Lead Product and the AstraZeneca Collaboration Products. The term of performance is approximately five years. The amounts allocated to the four performance obligations for the material rights to acquire a development license and the four performance obligations for commercial licenses for the AstraZeneca Collaboration Products will be recognized upon exercise of the specific material right and delivery of each of the development licenses. As of June 30, 2021, there was $21.8 million of aggregate transaction price allocated to remaining performance obligations.

Additionally, the Company evaluated payments required to be made between both parties as a result of the shared development costs of the AstraZeneca Lead Product and the two AstraZeneca Collaboration Products for which the Company has a co-development option. The Company will classify payments made as a reduction of revenue and will classify payments received as revenue in the period they are earned.

Under the AstraZeneca Agreements, the Company is eligible to receive various research, development, commercial and sales milestones. There is uncertainty that the events to obtain the research and development milestones will be achieved given the nature of clinical development and the stage of the Company’s technology. The Company has thus determined that all research and development milestones, other than the phase 1 initiation milestone achieved in December 2017 and included in the impact of adoption of ASC 606, will be constrained until it is deemed probable that a significant revenue reversal will not occur.

On March 29, 2021, the Company and AstraZeneca entered into (1) Amendment No. 1 to the Non-exclusive Anticalin® Platform License Agreement dated May 2, 2017 and (2) Amendment No. 2 to the License and Collaboration Agreement dated May 2, 2017, as previously amended by Amendment No. 1 dated September 14, 2020, collectively, the Amended Collaboration Agreement. Under the Amended Collaboration Agreement, the parties agreed to restructure certain commercial economics for the AZD1402/PRS-060 program by increasing potential sales milestones and reducing potential sales royalties, while fundamentally maintaining the overall value split between AstraZeneca and the Company.

In connection with the Amended Collaboration Agreement, the Company and AstraZeneca entered into a subscription agreement, or the AstraZeneca Subscription Agreement, pursuant to which the Company agreed to issue to AstraZeneca, and AstraZeneca agreed to acquire from the Company, 3,584,230 shares of the Company’s common stock for a total purchase price of $10.0 million, or $2.79 per share, in a private placement transaction pursuant to Section 4(a)(2) of the Securities Act. The
19


AstraZeneca Subscription Agreement closed on April 1, 2021 and includes a requirement that the Company file a registration statement to register the resale of the shares issued to AstraZeneca within 60 calendar days of the issuance of the shares. The Company assessed the payment under ASC 606 and concluded that the fair value of the shares on a per share basis was $2.60 per share as of the transaction date. This resulted in a premium paid for the shares of $0.7 million, which was added to the deferred revenue balance and will be recognized over time in line with our revenue recognition pattern for all remaining performance obligations.

Also in March 2021, the Company earned a $13.0 million milestone from AstraZeneca related to the initiation of the phase 2a study for PRS-060/AZD1402. The Company assessed the milestone payment under ASC 606 and determined that there no longer existed a constraint on the milestone as the performance obligation related to the phase 2a study was fully satisfied. Therefore, the Company realized the full $13.0 million as milestone revenue during the quarter ended March 31, 2021.

As of June 30, 2021, there were $1.0 million and $18.1 million of current and non-current deferred revenue, respectively, related to the AstraZeneca Agreements.

The Company incurred $1.6 million of third-party success fees to obtain the contract with AstraZeneca. Upon adoption of ASC 606, the Company capitalized $1.1 million in accordance with ASC 340. As of June 30, 2021, the remaining balance of the asset recognized from transaction costs to obtain the AstraZeneca contract was $0.7 million. Amortization during the three and six months ended June 30, 2021 and 2020 was de minimis.

Servier

On January 4, 2017, the Company entered into a license and collaboration agreement, or Servier Collaboration Agreement, and a non-exclusive Anticalin platform license agreement, or Servier Platform License, and together with the Servier Collaboration Agreement, the Servier Agreements, with Les Laboratoires Servier and Institut de Recherches Internationales Servier, or Servier, pursuant to which the Company and Servier agreed to initially pursue five bispecific therapeutic programs.

Five committed programs were initially defined, which may combine antibodies from the Servier portfolio with one or more Anticalin proteins based on the Company’s proprietary platform to generate innovative IO bispecific drug candidates, or the Collaboration Products. The collaboration may be expanded by up to three additional therapeutic programs. The Company had the option to co-develop and retain commercial rights in the United States for PRS-332, the initial lead program under the collaboration, or the Initial Lead, and has a similar option on up to three additional programs, or the Co-Development Collaboration Products, while Servier will be responsible for development and commercialization of the other programs worldwide, or the Servier Worldwide Collaboration Products. Each party is responsible for an agreed upon percentage of shared costs, as set forth in the budget for the collaboration plan, and as further discussed below.

The Co-Development Collaboration Products may be jointly developed, according to a collaboration plan, through marketing approval from the U.S. Food and Drug Administration or the European Medicines Agency. Servier Worldwide Collaboration Products may be jointly developed, according to a collaboration plan, through specified preclinical activities, at which point Servier becomes responsible for further development of the Collaboration Product.

At inception, Servier was granted the following licenses: (i) development license for the Initial Lead, (ii) commercial license for the Initial Lead, (iii) individual research licenses for each of the four Collaboration Products, and (iv) individual non-exclusive platform technology licenses for the Initial Lead and for each of the four Collaboration Products. Upon achievement of certain development activities, specified by the collaboration for each Servier Agreement, Servier will be granted a development license and a commercial license. For the Initial Lead and the Co-Development Collaboration Products, the licenses granted are with respect to the entire world except for the United States. For Servier Worldwide Collaboration Products, the licenses granted are with respect to the entire world.

The Servier Agreements are managed on an overall basis by a joint executive committee, or JEC, formed by an equal number of members from the Company and Servier. Decisions by the JEC will be made by consensus; however, in the event of a disagreement, each party will have final-decision making authority as it relates to the applicable territory in which such party has commercialization rights for the applicable product. In addition to the JEC, the Servier Collaboration Agreement requires the participation of both parties on: (i) a JSC, (ii) a JDC, (iii) a joint intellectual property committee, or JIPC, and (iv) a joint research committee, or JRC. The responsibilities of these committees vary, depending on the stage of development and commercialization of the Collaboration Products.

 
For the Initial Lead and Co-Development Collaboration Products, the Company and Servier are responsible for an agreed upon percent of the shared costs required to develop the products through commercialization. In the event that the Company fails to
20


exercise its option to co-develop the Co-Development Collaboration Products, Servier has the right to continue with the development and will be responsible for all costs required to develop the products through commercialization.

Under the Servier Agreements, the Company received an upfront, non-refundable payment of €30.0 million (approximately $32.0 million). In addition, the Company is eligible to receive research, development, commercial and sales milestone payments as well as tiered royalties up to low double digits on the sales of commercialized products in the Servier territories. The Company achieved two preclinical milestones under the program, one in December 2018 for €0.5 million (approximately $0.6 million) and another in February 2019 for €1.5 million (approximately $1.7 million), both of which became billable on their respective achievement dates.

The initial research collaboration term, as it relates to the Initial Lead and Collaboration Products, shall continue for three years from the effective date of the Servier agreements and may be mutually extended for two one-year terms consecutively applied.

The term of each Servier Agreement ends upon the expiration of all of Servier’s payment obligations under such Servier Agreement. The Servier Agreements may be terminated by Servier for convenience beginning 12 months after their effective date upon 180 days’ notice. The Servier Agreements may also be terminated by Servier or the Company for material breach upon 90 days’ or 120 days’ notice under the Servier Collaboration Agreement and the Servier Platform License, respectively, provided that the applicable party has not cured such breach by the applicable 90-day or 120-day permitted cure period, and dispute resolution procedures specified in the applicable Servier Agreement have been followed. The Servier Agreements may also be terminated due to the other party’s insolvency or for a safety issue and may in certain instances be terminated on a product-by-product and/or country-by-country basis. The Servier Platform License will terminate upon termination of the Servier Collaboration Agreement, on a product-by-product and/or country-by-country basis.

As the Company and Servier are considered to be active participants in the Servier Agreements and are exposed to significant risks and rewards, certain units of account within the Servier Agreements are within the scope of ASC 808. The arrangement with Servier provides for the transfer of the following goods and services: (i) five non-exclusive platform technology licenses, a development license, a commercial license and research and development services for the Initial Lead, (ii) participation on each of the committees, (iii) four research licenses for Collaboration Products, and (iv) research and development services for the Collaboration Products. Additionally, as the development and commercial licenses on the four Collaboration Products may be granted at a discount in the future, the Company determined such discounts should be assessed as material rights at inception.

Management evaluated all of the promised goods or services within the contract and determined which goods and services
were separate performance obligations. The Company determined that the licenses granted, at the inception of the Servier collaboration, should be combined with the research and development services to be provided for the Initial Lead and Collaboration Products, over the term of the Servier Agreements, as such licenses are not capable of being distinct. A third party would not be able to provide the research and development services, due to the specific nature of the intellectual property and knowledge required to perform the services, and Servier could not benefit from the licenses without the corresponding services. The Company determined that the participation on the various committees was distinct as the services could be performed by an outside party.

As a result, management concluded that there were 10 performance obligations at the inception of the Servier Agreements. The following performance obligations are within the scope of ASC 808: (i) combined performance obligation comprised of a non-exclusive platform technology license, commercial license, development license and research and development services for the Initial Lead, (ii) two separate performance obligations each comprised of a combined non-exclusive platform technology license, research license and research and development services for each Co-Development Collaboration Product, (iii) one performance obligation comprised of participation in the various governance committees, and (iv) two combined performance obligations comprised of the development and commercial licenses granted for the Co-Development Collaboration Products (and corresponding discounts) upon the achievement of specified preclinical activities, resulting in material rights. Revenue recognized associated with these performance obligations are presented as Collaboration Revenue within the Statement of Operations. The following performance obligations are within the scope of ASC 606: (i) two separate performance obligations each comprised of a combined non-exclusive platform technology license, research license and research and development services for each Servier Worldwide Collaboration Product, and (ii) two combined performance obligations comprised of the development and commercial licenses granted for the Servier Worldwide Collaboration Products (and corresponding discounts) upon the achievement of specified preclinical activities, resulting in material rights. Revenue recognized associated with these performance obligations are presented as Customer Revenue within the Statement of Operations.

The Company allocated consideration to the performance obligations based on the relative proportion of their standalone selling prices. The Company developed its standalone selling prices for licenses by applying a risk adjusted, net present value, estimate
21


of future potential cash flows approach, which included the cost of obtaining research and development services at arm’s length from a third-party provider, as well as internal full-time equivalent costs to support these services.

The Company developed its estimate of standalone selling price for committee participation by using management’s estimate of the anticipated participation hours multiplied by a market rate for comparable participants.

The Company developed its estimate of standalone selling price for the material rights granted on the development and commercial licenses granted for the Collaboration Products by probability weighting multiple cash flow scenarios using the income approach.

The transaction price at inception is comprised of the fixed upfront fee of €30.0 million (approximately $32.0 million) and was allocated to the performance obligations based on the relative proportion of their standalone selling prices.

The amounts allocated to the performance obligation for the Initial Lead and the four performance obligations for the four research and development licenses for Collaboration Products will be recognized on a proportional performance basis as the activities are conducted over the life of the arrangement. The term of the performance at inception of the Servier Agreements for the Initial Lead and each of the Co-Development Collaboration Products may be through approval of certain regulatory bodies; a period which could be many years. The term of the performance for each of the other two Servier Worldwide Collaboration Products is through the initial research and collaboration term, plus potential extensions. The amounts allocated to the performance obligation for participation on each of the committees will be recognized on a straight-line basis over the anticipated performance period over the entirety of the arrangement with Servier. The amounts allocated to the four performance obligations for the material rights to acquire development and commercial licenses for the Co-Development Collaboration Products are granted in the future will be recognized over time upon delivery of each of the licenses through marketing approval. The amounts allocated to the four performance obligations for the material rights to acquire development and commercial licenses for the Servier Developed Collaboration Products are granted in the future will be recognized upon delivery of each of the licenses. As of June 30, 2021, there was $10.9 million of aggregate transaction price allocated to remaining performance obligations.

Additionally, the Company evaluated payments required to be made between both parties as a result of the shared development costs of the Initial Lead and Collaboration Products. The Company will classify payments made as a reduction of revenue and will classify payments received as revenue, in the period they are earned.

Under the Servier Agreements, the Company is eligible to receive various research, development, commercial and sales milestones. There is uncertainty that the events to obtain the research and development milestones will be achieved given the nature of clinical development and the stage of the Company’s technology. The Company has thus determined that all research and development milestones will be constrained until it is deemed probable that a significant revenue reversal will not occur.

In September 2019, Servier notified the Company of its decision to discontinue co-development of PRS-332, a PD-1-LAG-3 bispecific that served as the initial development program under the Pieris-Servier alliance, for strategic reasons. The Company does not presently intend to continue development of PRS-332 but retains full rights to advance the development and commercialization of the product on a world-wide basis in the future.

In February 2020, the research term was extended for another 12 months. The Company has updated the transaction price for the extension for revenue recognition purposes and allocated it ratably over all unsatisfied performance obligations. In March 2020, Servier notified the Company of its decision to discontinue co-development of two earlier preclinical stage programs for strategic reasons based upon an extensive portfolio review. The notification required a 60-day period to complete remaining obligations on the programs; however, the Company determined that the material rights to acquire development and commercial licenses for one Co-Development Collaboration Product and for one Servier Developed Collaboration Products lapsed in March 2020 and recognized as revenue $7.1 million of previously deferred revenue associated with these material rights during the three-month period ended March 31, 2020. The parties continue to advance the development of two preclinical programs: PRS-344, a 4-1BB/PD-L1 bispecific designed as a co-development program, and PRS-352, which addresses undisclosed targets and for which Servier has worldwide rights.

In February 2021, the research term was extended for another 12 months.

As of June 30, 2021, there were $6.5 million and $4.4 million of current and non-current deferred revenue, respectively, related to the Servier Agreements.

22


The Company incurred costs to obtain the contract with Servier. Upon adoption of ASC 606, the Company capitalized $0.5 million of third-party service fees in accordance with ASC 340. As of June 30, 2021, the remaining balance of the asset recognized from costs to obtain the Servier contract was $0.1 million. Amortization during the three and six months ended June 30, 2021 was de minimis. Amortization during the three and six months ended June 30, 2020 was de minimis and $0.1 million, respectively.

Contract Balances
The Company receives payments from its collaboration partners based on payments established in each contract. Upfront payments and fees are recorded as deferred revenue upon receipt or when due until such time as the Company satisfies its performance obligations under each arrangement. A contract asset is a conditional right to consideration in exchange for goods or services that the Company has transferred to a customer. Amounts are recorded as accounts receivable when the Company’s right is unconditional.

Additions to deferred revenue were $26.1 million and $30.1 million during the three and six months ended June 30, 2021, respectively. Reductions to deferred revenue were $0.7 million and $1.5 million for the three and six months ended June 30, 2021, respectively.

4.    Grant Income

One of the Company's proprietary respiratory assets is PRS-220, an oral inhaled Anticalin protein targeting connective tissue growth factor, or CTGF, and it is being developed as a local treatment for idiopathic pulmonary fibrosis. In June 2021, the Company was selected to receive a €14.2 million (approximately $17.0 million) grant from the Bavarian Ministry of Economic Affairs, Regional Development and Energy (the Bavarian Grant) supporting research and development for post-acute sequelae of SARS-CoV-2 infection (PASC) pulmonary fibrosis, or PASC-PF, also known as post-COVID-19 syndrome pulmonary fibrosis, or “long COVID”.

The Bavarian Grant provides partial reimbursement for qualifying research and development activities on PRS-220, including drug manufacturing costs, activities and costs to support an IND filing, and phase 1 clinical trials costs. The Bavarian Grant provides reimbursement of qualifying costs incurred through August 2023, which follows the expected development timeline of this program. Qualifying costs incurred may exceed the annual grant funding thresholds. If the Company receives any proceeds from the sale of or licensing income from PRS-220, the funds available for reimbursement will be reduced proportionally if they are obtained prior to August 2023. The Company is required to communicate such proceeds in each case with the request to draw-down the funds.

5.    Cash, cash equivalents and investments
As of June 30, 2021 and December 31, 2020, cash equivalents were $91.8 million and $64.0 million, respectively, and are comprised of money market accounts, all of which are Level 1 investments. The Company did not hold any Level 2 or 3 investments as of June 30, 2021 and did not have any transfers of investment between levels for the three and six months ended June 30, 2021.
The Company did not record any realized gains or losses from the maturity of available-for-sale securities during the three and six months ended June 30, 2021. The Company recorded de minimis realized gains and a realized gain of $0.2 million from the maturity of available-for-sale securities for the three and six months ended June 30, 2020, respectively.

6.    Property and equipment, net
Property and equipment are summarized as follows (in thousands):
 June 30,December 31,
 20212020
Laboratory furniture and equipment$11,043 $11,188 
Office furniture and equipment2,056 2,120 
Computer equipment410 394 
Leasehold improvements13,723 14,159 
Property and equipment, cost27,232 27,861 
Accumulated depreciation(6,859)(5,815)
Property and equipment, net$20,373 $22,046 
23



7.    Accrued Expenses
Accrued expenses and other current liabilities consisted of the following (in thousands):
 June 30,December 31,
 20212020
Accrued accounts payable$4,215 $1,220 
Collaboration cost-sharing obligation 3,830  
Research and development fees2,813 2,001 
Compensation expense2,251 2,759 
Accrued license obligations1,653 358 
Lease liabilities959 1,030 
Audit and tax fees189 128 
Other current liabilities173 235 
Total $16,083 $7,731 

8.    Net Loss per Share

Basic net loss per share is calculated by dividing net income loss by the weighted average shares outstanding during the period, without consideration for common stock equivalents. Diluted net loss per share is calculated by adjusting weighted average shares outstanding for the dilutive effect of common stock equivalents outstanding for the period, determined using the treasury-stock and if-converted methods. For purposes of the diluted net loss per share calculation, preferred stock, stock options and warrants are considered to be common stock equivalents but have been excluded from the calculation of diluted net loss per share, as their effect would be anti-dilutive for all periods presented. Therefore, basic and diluted net loss per share were the same for all periods presented.

As of June 30, 2021 and 2020, and as calculated using the treasury stock method, approximately 36.5 million and 37.3 million of weighted average shares, respectively, were excluded from the calculation of diluted weighted average shares outstanding as their effect was antidilutive.

9.    Stockholders’ Equity
The Company had 300,000,000 shares authorized and 66,679,223 and 56,002,815 shares of common stock issued and outstanding as of June 30, 2021 and December 31, 2020, respectively, with a par value of $0.001 per share.
The Company had 10,000,000 shares authorized and 15,617 shares of preferred stock issued and outstanding as of June 30, 2021. The Company had 10,000,000 shares authorized and 14,429 shares of preferred stock issued and outstanding as of December 31, 2020. Preferred stock has a par value of $0.001 per share, and consists of the following:
Series A Convertible, 85 and 2,907 shares issued and outstanding at June 30, 2021 and December 31, 2020, respectively.
Series B Convertible, 4,026 and 5,000 shares issued and outstanding at June 30, 2021 and December 31, 2020, respectively.
Series C Convertible, 3,506 and 3,522 shares issued and outstanding at June 30, 2021 and December 31, 2020, respectively.
Series D Convertible, 3,000 shares issued and outstanding at June 30, 2021 and December 31, 2020, respectively.
Series E Convertible, 5,000 and 0 shares issued and outstanding at June 30, 2021 and December 31, 2020, respectively.
2020 Employee, Director and Consultant Equity Incentive Plan

At the 2020 Annual Meeting of Stockholders, the Company's stockholders approved the 2020 Employee, Director and Consultant Equity Incentive Plan, or the 2020 Plan. The 2020 Plan permits the Company to issue up to 3,500,000 shares of common stock pursuant to awards granted under the 2020 Plan. Upon approval of the 2020 Plan, the 2019 Employee, Director and Consultant Equity Incentive Plan, or the 2019 Plan, was terminated; all unissued options were canceled and no additional
24


awards will be made thereunder. All outstanding awards under the 2019 Plan will remain in effect and any awards forfeited from the outstanding awards will be allocated back into the 2020 Plan. There were approximately 1,579,678 shares remaining and available for grant under the 2019 Plan that terminated upon original approval of the 2020 Plan.

At the 2021 Annual Meeting of Stockholders, held on June 25, 2021, the Company's stockholders approved the first amendment to the 2020 Plan to add 2,250,000 shares for issuance under the 2020 Plan.

Series D Preferred Stock Conversion

On March 31, 2020, the Company and certain entities affiliated with Biotechnology Value Fund, L.P., or BVF, entered into an exchange agreement pursuant to which, on April 1, 2020, BVF exchanged an aggregate of 3,000,000 shares of the Company’s common stock owned by BVF for an aggregate of 3,000 shares of Series D Preferred Stock. The Company designated 3,000 shares of its authorized and unissued preferred stock as Series D Preferred Stock and filed a Certificate of Designation of Series D Convertible Preferred Stock of Pieris Pharmaceuticals, Inc. with the Nevada Secretary of State.

Series E Preferred Stock Conversion

On May 20, 2021, the Company and certain entities affiliated with BVF entered into an exchange agreement pursuant to which, BVF exchanged an aggregate of 5,000,000 shares of the Company’s common stock owned by BVF for an aggregate of 5,000 shares of Series E Preferred Stock. The Company designated 5,000 shares of its authorized and unissued preferred stock as Series E Preferred Stock and filed a Certificate of Designation of Series E Convertible Preferred Stock of Pieris Pharmaceuticals, Inc., or the Series E Certificate of Designation, with the Nevada Secretary of State.

As described below, the Series E Preferred Stock has substantially the same terms as the Company’s Series D Convertible Preferred Stock, par value $0.001 per share, issued in April 2020; Series C Convertible Preferred Stock, par value $0.001 per share, issued in November 2019; Series B Convertible Preferred Stock, par value $0.001 per share, issued in January 2019; and Series A Convertible Preferred Stock, par value $0.001 per share, issued in June 2016, all currently held by entities affiliated with BVF.

Each share of Series E Preferred Stock is convertible into 1,000 shares of Common Stock (subject to adjustment as provided in the Series E Certificate of Designation) at any time at the option of the holder, provided that the holder is prohibited from converting the Series E Preferred Stock into shares of Common Stock if, as a result of such conversion, the holder, together with its affiliates, would own more than 9.99% of the total number of shares of Common Stock then issued and outstanding, or the Beneficial Ownership Limitation. The holder may reset the Beneficial Ownership Limitation to a higher or lower number (not to exceed 19.99% of the total number of Common Shares issued and outstanding immediately after giving effect to a conversion) upon providing written notice to the Company. Any such notice providing for an increase to the Beneficial Ownership Limitation will be effective 61 days after delivery to the Company. In the event of the Company’s liquidation, dissolution, or winding up, subject to the rights of holders of Senior Securities (defined below), holders of Series E Preferred Stock are entitled to receive a payment equal to $0.001 per share of Series E Preferred Stock before any proceeds are distributed to the holders of Common Stock and Junior Securities (defined below) and pari passu with any distributions to the holders of the Series A Preferred Stock, Series B Preferred Stock, Series C Preferred Stock and Series D Preferred Stock, plus an additional amount equal to any dividends declared but unpaid on such shares. However, if the assets of the Company are insufficient to comply with the preceding sentence, then all remaining assets of the Company shall be distributed ratably to holders of the shares of the Series E Preferred Stock and Parity Securities (defined below). Shares of Series E Preferred Stock generally have no voting rights, except as required by law and except that the consent of holders of a majority of the then outstanding Series E Preferred Stock is required to amend the terms of the Series E Certificate of Designation. Holders of Series E Preferred Stock are entitled to receive any dividends payable to holders of Common Stock, and rank:
senior to all of the Common Stock;
senior to any class or series of capital stock of the Company created after the designation of the Series E Preferred Stock specifically ranking by its terms junior to the Series E Preferred Stock, or the Junior Securities;
on parity with all shares of Series A Preferred Stock, Series B Preferred Stock, Series C Preferred Stock, Series D Preferred Stock and any class or series of capital stock of the Company created after the designation of the Series E Preferred Stock specifically ranking by its terms on parity with the Series E Preferred Stock, or the Parity Securities; and
junior to any class or series of capital stock of the Company created after the designation of the Series E Preferred Stock specifically ranking by its terms senior to the Series E Preferred Stock, or the Senior Securities,
25


in each case, as to distributions of assets upon the Company’s liquidation, dissolution or winding up whether voluntarily or involuntarily and/or the right to receive dividends.
Open Market Sales Agreement
In August 2019, the Company entered into a sales agreement pursuant to which the Company may offer and sell shares of its common stock, from time to time, up to an aggregate amount of gross sales proceeds of $50.0 million through the ATM Program under a shelf registration statement on Form S-3. Through June 30, 2021, the Company has sold $22.7 million of common shares under the ATM Program of which 3.0 million shares were sold during the quarter ending June 30, 2021 at an average stock price of $4.24.
10.    Leases
The Company currently leases office space in Boston, Massachusetts. In August 2015, the Company entered into a sublease to lease approximately 3,950 square feet. The sublease originally expired on February 27, 2022 or such earlier date pursuant to the termination provisions of the sublease. In July 2021, the Company extended the lease for this office space for an additional 10 months through December 31, 2022.
The Company also leased approximately 19,000 square feet of office and laboratory space in Freising, Germany under four agreements, or the Freising Leases, including three leases for space on three floors of the same building and a letter agreement for additional conference room space within the building. The Freising Leases expired on March 31, 2020.

In October 2018, Pieris GmbH entered into a new lease for office and laboratory space located in Hallbergmoos, Germany, or the Hallbergmoos Lease. Pieris GmbH moved its operations, formerly conducted in Freising, Germany, to the Hallbergmoos facility in February 2020.

Under the Hallbergmoos Lease, Pieris GmbH will rent approximately 105,000 square feet, of which approximately 98,400 square feet were delivered by the lessor in February 2020 and approximately 5,100 square feet were delivered by the lessor in May 2020. An additional approximately 22,300 square feet is expected to be delivered by the lessor by October 2024. Pieris GmbH has a first right of refusal to lease an additional approximate 13,400 square feet.

The Hallbergmoos Lease provides for an initial rental term of 12.5 years which commenced in February 2020 when the leased property was delivered to Pieris GmbH. Pieris GmbH also has an option to extend the Hallbergmoos Lease for two additional 60-month periods. The Company is not reasonably certain to exercise the option to extend the lease expiration beyond its current expiration date. Pieris GmbH may sublease space within the leased property with lessor’s consent, which may not be unreasonably withheld.

Monthly base rent for the initial 105,000 square feet of the leased property, including parking spaces, will total approximately $0.2 million per month, which amount shall be adjusted starting on the second anniversary of the commencement date by an amount equal to the German consumer price index. In addition to the base rent, Pieris GmbH is also responsible for certain administrative and operational costs in accordance with the Hallbergmoos Lease. Pieris GmbH provided a security deposit of $0.8 million as required by the Hallbergmoos Lease. The Company will serve as a guarantor for the Hallbergmoos Lease.

The Hallbergmoos Lease included $11.5 million of tenant improvements allowance for normal tenant improvements, for which construction began in March 2019. The Company capitalized the leasehold incentives which are included in Property and equipment, net on the Condensed Consolidated Balance Sheet and are amortized on a straight-line basis over the shorter of the useful life or the remaining lease term. The lease incentive allowance was also factored in as a reduction to the right-of-use asset upon the adoption of ASC 842.

The following table summarizes operating lease costs included in operating expenses (in thousands):
 Three Months Ended June 30,Six Months Ended June 30,
2021202020212020
Operating lease costs$379 $347 $744 $751 
Variable lease costs (1)181 156 364 344 
Total lease cost$560 $503 $1,108 $1,095 
(1) Variable lease costs include certain additional charges for operating costs, including insurance, maintenance, taxes, utilities, and other costs incurred, which are billed based on both usage and as a percentage of the Company’s share of total square footage.
26


The following table summarizes the weighted-average remaining lease term and discount rate:
 As of June 30, 2021
Weighted-average remaining lease term (years)11.0
Weighted-average discount rate10.5 %
Cash paid for amounts included in the measurement of the lease liabilities was $0.6 million and $1.3 million, respectively, for the three and six months ended June 30, 2021. Cash paid for amounts included in the measurement of the lease liabilities was $0.6 million and $0.9 million, respectively, for the three and six months ended June 30, 2020.
As of June 30, 2021, the maturities of the Company’s operating lease liabilities and future minimum lease payments were as follows (in thousands):
 Total
2021$1,283 
20222,380 
20232,346 
20242,346 
20252,346 
Thereafter15,443 
Total undiscounted lease payments26,144 
Less: present value adjustment(10,224)
Present value of lease liabilities$15,920 
27



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

The interim financial statements and this Management’s Discussion and Analysis of Financial Condition and Results of Operations should be read in conjunction with the financial statements and notes thereto for the year ended December 31, 2020, and the related Management’s Discussion and Analysis of Financial Condition and Results of Operations, both of which are contained in our Annual Report on Form 10-K for the fiscal year ended December 31, 2020, filed with the SEC on March 31, 2021. In addition to historical information, this discussion and analysis contains forward-looking statements that involve risks, uncertainties and assumptions. Our actual results may differ materially from those anticipated in these forward-looking statements as a result of certain factors, including but not limited to those set forth under the caption “Risk Factors” in the Annual Report on Form 10-K for the fiscal year ended December 31, 2020.

As used in this Quarterly Report on Form 10-Q, unless the context indicates or otherwise requires, “our Company”, “the Company”, “Pieris”, “we”, “us” and “our” refer to Pieris Pharmaceuticals, Inc., a Nevada corporation, and its consolidated subsidiaries.

We have registered trademarks for Pieris, Anticalin and others. All other trademarks, trade names and service marks included in this Quarterly Report on Form 10-Q are the property of their respective owners. Use or display by us of other parties’ trademarks, trade dress or products is not intended to and does not imply a relationship with, or endorsements or sponsorship of, us by the trademark or trade dress owner.
Overview
We are a clinical-stage biotechnology company that discovers and develops Anticalin-based drugs to target validated disease pathways in unique and transformative ways. Our clinical pipeline includes an inhaled IL-4Rα antagonist Anticalin protein to treat moderate-to-severe asthma and an IO bispecific targeting 4-1BB and HER2. Proprietary to us, Anticalin proteins are a novel class of therapeutics validated in the clinic and through partnerships with leading pharmaceutical companies. Our core Anticalin technology and platform were developed in Germany, and we have collaborations with major multi-national pharmaceutical companies. In particular, we have alliances with AstraZeneca and Genentech to treat respiratory diseases, with Genentech also in ophthalmology, and partnerships with Servier and Seagen, both in IO. Our development programs include:

PRS-060/AZD1402, our lead respiratory program partnered with AstraZeneca, is a drug candidate that antagonizes IL-4Rα, thereby inhibiting the downstream action of IL-4 and IL-13, two cytokines known to be key mediators in the inflammatory cascade that drive the pathogenesis of asthma and other inflammatory diseases.

Four respiratory programs included in the AstraZeneca alliance beyond PRS-060/AZD1402, the targets and disease areas of which are undisclosed. We retain co-development and co-commercialization rights to two out of these four programs.

Our lead fully proprietary respiratory asset PRS-220, an oral inhaled Anticalin protein targeting connective tissue growth factor, or CTGF, is being developed as a local treatment for idiopathic pulmonary fibrosis and has passed drug candidate nomination stage. We were selected to receive a €14.2 million grant from the Bavarian Ministry of Economic Affairs, Regional Development and Energy supporting research and development of the program for post-acute sequelae of SARS-CoV-2 infection (PASC) pulmonary fibrosis, or PASC-PF, also known as post-COVID-19 syndrome pulmonary fibrosis, or “long COVID”.

We have also entered into a multi-program research collaboration and license agreement with Genentech, a member of the Roche Group, to discover, develop and commercialize locally delivered respiratory and ophthalmology therapies.

Cinrebafusp alfa, our lead IO program, is a fusion protein, comprising a HER2-targeting antibody genetically linked to 4-1BB-targeting Anticalin proteins. Cinrebafusp alfa is designed to drive tumor localized T cell activation through tumor-targeted drug clustering mediated by HER2 expressed on tumor cells. This program was the first bispecific T cell co-stimulatory agonist to enter clinical development.

PRS-344, a bispecific antibody-Anticalin fusion protein comprising an PD-L1-targeting antibody genetically fused to Anticalin proteins specific for 4-1BB. PRS-344 is being developed as part of our IO collaboration with Servier.

We are also developing additional IO drug candidates beyond cinrebafusp alfa and PRS-344 that are multi-specific Anticalin-based fusion proteins designed to engage immunomodulatory targets, comprising a variety of
28


multifunctional biotherapeutics. Other IO drug candidates are being developed as part of our collaboration with Servier and Seagen.

We are supporting IND-readiness for PRS-342, a 4-1BB/GPC3 bispecific that we have exclusively licensed to Boston Pharmaceuticals, who will oversee future development of that asset.

Our programs are in varying stages:

PRS-060/AZD1402 was tested in a nebulized formulation in 54 healthy volunteers at nominal dose levels ranging from 0.25 mg to 400 mg in a phase 1 single ascending dose study. Data from that study were presented at the American Thoracic Society International Conference in May 2019 showing that PRS-060/AZD1402 was well-tolerated when given as single inhaled or intravenous doses to healthy volunteers and there was systemic target engagement (as measured by pSTAT6 inhibition). We presented interim data from the PRS-060/AZD1402 phase 1 multiple ascending dose study at the European Respiratory Society International Congress in October 2019 and reported that PRS-060/AZD1402 was safe and well-tolerated at all doses, led to a statistically significant reduction in FeNO, a validated biomarker for eosinophilic airway inflammation, and showed dose-dependent systemic target engagement in patients with mild asthma and elevated levels of FeNO (≥ 35ppb).

In addition to Ukrainian regulatory approval, AstraZeneca has also recently received ethics approval and regulatory acknowledgement for the phase 2a study of PRS-060/AZD1402 in Australia and is actively recruiting patients in both countries. Following a COVID-19-related inventory challenge, the first patient has been dosed in Ukraine and screening for the study is ongoing in both Ukraine and Australia. As part of the phase 2a initiation, we earned a $13.0 million milestone payment from AstraZeneca. The phase 2a study is a two-part, multi-center, placebo-controlled clinical study of PRS-060/AZD1402 that will evaluate PRS-060/AZD1402 at up to three dose levels using a dry powder formulation administered twice daily. The first part of the study will assess the safety and pharmacokinetics of the dry powder formulation in approximately 45 moderate controlled asthmatics. The second part of the study will assess the efficacy, safety and pharmacokinetics of PRS-060/AZD1402 over four weeks in approximately 360 moderate uncontrolled asthmatics with blood eosinophil count of ≥ 150 cells/μL and FeNO ≥ 25 ppb at screening with FEV1 improvement as the primary endpoint. This study evaluating PRS-060/AZD1402, which is being developed for the treatment of moderate-to-severe asthma, is being sponsored, funded and delivered by AstraZeneca. Upon completion of that study, Pieris will have the options to co-develop and, subsequently, co-commercialize PRS-060/AZD1402 in the United States.

Our additional respiratory programs, both partnered and proprietary, are in the discovery stage. AstraZeneca has taken advantage of all available potential new project starts envisioned in the alliance and all four respiratory programs are ongoing. We retain co-development and co-commercialization rights to two out of the four programs beyond PRS-060/AZD1402.

The PRS-220 program is currently in the IND-enabling stage, with clinical development expected to begin in 2022.

In January 2021, the FDA lifted the partial clinical hold of the phase 1 studies of cinrebafusp alfa. Cinrebafusp alfa was placed on partial clinical hold by the FDA in July 2020 while we conducted an additional in-use stability and compatibility study requested by the agency. Treatment of enrolled patients continued, although no new patients were enrolled pending resolution of the partial hold. We completed the in-use studies deemed necessary in connection with the partial clinical hold. As part of the completed studies that supported a robust process for administration of cinrebafusp alfa in the clinical setting, we have optimized the level of an existing excipient to enhance the stability of cinrebafusp alfa under prescribed as well as stressed conditions that could occur in preparation of the drug candidate for patient administration in the real-world clinical setting.

We are now preparing a two-arm phase 2 study for cinrebafusp alfa in gastric cancer that will begin this summer. Supported by additional data we presented from the phase 1 monotherapy study of cinrebafusp alfa in an oral presentation session at the American Association for Cancer Research Virtual Congress, or AACR, in April 2021, the first arm of the phase 2 study will include the combination with ramucirumab and paclitaxel in HER2-high gastric cancer, while the second arm will be in combination with tucatinib in HER2-low gastric cancer. Collaboration partners Lilly and Seagen will supply ramucirumab and tucatinib, respectively. Go/No-Go criteria for advancement of this program will evaluate a composite of measures, including a minimum target of 50% ORR in the HER2-high arm and a minimum target of 40% ORR in the HER2-low arm, duration of response, and safety. We expect to report results from both study arms next year. In June 2021, FDA granted orphan drug designation to cinrebafusp alfa for the treatment of HER2-high and HER2-low expressing gastric cancers.
29


The supporting data presented at AACR included an evaluation of 78 patients who had been enrolled in the monotherapy study as of the February 2021 cutoff date, including four additional patients enrolled in the active dose cohorts (≥2.5 mg/kg) since the data were presented at the European Society for Medical Oncology, or ESMO, Virtual Congress in September 2020. Out of 42 response-evaluable patients at the time of the data cutoff of February 25, 2021, according to RECIST 1.1, one patient with stage 4 rectal adenocarcinoma achieved a confirmed complete response at the 18 mg/kg Q2W dose (cohort 13b), four patients achieved a partial response (three at the 8 mg/kg Q2W dose (cohort 11b) and one at the 18 mg/kg Q2W dose (cohort 13b)), and stable disease was observed in 17 patients as best response out of 42 evaluable patients across the predicted active dose ranges (cohorts 9-13b), translating to an ORR of 12% and a DCR of 52%. Consistent with the MOA of cinrebafusp alfa, dose-dependent immune activation was demonstrated by showing an increase in CD8+, T cell, NK cells and cytotoxic activity in the tumor microenvironment and an increase of soluble 4-1BB in the blood, indicating target engagement of 4-1BB and activation of immune cells. Cinrebafusp alfa demonstrated durable anti-tumor activity in a heavily pre-treated patient population. Additionally, clinical benefit was observed in patients with “cold” tumors as well as those with low HER2 expression who were enrolled into the study on the basis of archived HER2-status and were later re-assessed on the basis of a pre-treatment biopsy. Cinrebafusp alfa also showed an acceptable safety profile at all doses and schedules tested in the clinical study with no dose-limiting toxicities. The totality of response data generated in cohorts 11b (8 mg/kg Q2W) and 13b support the recommended phase 2 dose of a two-cycle loading dose of 18 mg/kg (Q2W), following by an 8 mg/kg dose (Q2W) in subsequent cycles.

The last update of the atezolizumab combination study of cinrebafusp alfa was presented at the ESMO Virtual Congress in September 2020. As of the July 2020 cutoff date, 41 patients had been enrolled and seven dose cohorts have been evaluated at a Q3W dosing schedule ranging from 0.05 mg/kg to 8 mg/kg in combination with a fixed 1200 mg dose of atezolizumab. In that trial, under RECIST 1.1, four patients achieved a confirmed partial response at active dose levels and an acceptable safety profile was observed at all doses and schedules tested in the clinical study.
For our additional IO drug candidates, we are conducting activities relating to candidate identification, optimization and preclinical evaluation.

We anticipate initiating a phase 1 study for PRS-344, a 4-1BB/PD-L1 bispecific, in 2021.
Servier has obtained in vivo proof of concept for PRS-352, an Anticalin-based bispecific beyond 4-1BB, triggering an undisclosed milestone payment to Pieris.Servier is responsible for further development of the program.
We achieved a key development milestone during 2020 for one of the programs in the Seagen collaboration, a bispecific tumor-targeted costimulatory agonist, triggering a $5.0 million milestone. We also handed the program over to Seagen, who is responsible for further advancement and funding of the asset. The program is one of up to three potential programs in the Seagen alliance, and we believe the achieved milestone further validates our approach and leadership in IO bispecifics, complementing the encouraging clinical data seen with cinrebafusp alfa.
We are supporting IND-readiness for PRS-342, a 4-1BB/GPC3 bispecific that we have exclusively licensed to Boston Pharmaceuticals, who will oversee future development of the asset.
Since inception, we have devoted nearly all of our efforts and resources to our research and development activities and have incurred significant net losses. For the three and six months ended June 30, 2021, we reported net losses of $15.5 million and $19.7 million, respectively. For the three and six months ended June 30, 2020, we reported net losses of $5.0 million and $8.5 million, respectively. As of June 30, 2021, we had an accumulated deficit of $231.1 million. We expect to continue incurring substantial losses for the next several years as we continue to develop our clinical and preclinical drug candidates and programs. Our operating expenses are comprised of research and development expenses and general and administrative expenses.
We have not generated any revenues from product sales to date and we do not expect to generate revenues from product sales for the foreseeable future. Our revenues for the three and six months ended June 30, 2021 and 2020 were from license and collaboration agreements with our partners.
A significant portion of our operations are conducted in countries other than the United States. Since we conduct our business in U.S. dollars, our main exposure, if any, results from changes in the exchange rates between the euro and the U.S. dollar. At each period end, we remeasure assets and liabilities to the functional currency of that entity (for example, U.S. dollar payables recorded by Pieris Pharmaceuticals GmbH). Remeasurement gains and losses are recorded in the statement of operations line item “Other income (expense), net.” All assets and liabilities denominated in euros are translated into U.S. dollars at the exchange rate on the balance sheet date. Revenues and expenses are translated at the weighted average rate during the period. Equity transactions are translated using historical exchange rates. All adjustments resulting from translating foreign currency financial statements into U.S. dollars are included in accumulated other comprehensive loss.
30


Key Financial Terms and Metrics
The following discussion summarizes the key factors our management believes are necessary for an understanding of our consolidated financial statements.
Revenues
We have not generated any revenues from product sales to date and we do not expect to generate revenues from product sales for the foreseeable future. Our revenues for the last two years have been primarily from the license and collaboration agreements with our partners.
The revenues from our partners have been comprised primarily of upfront payments, research and development services and milestone payments. For additional information about our revenue recognition policy, see “Note 2— Summary of Significant Accounting Policies.”
Research and Development Expenses
The process of researching and developing drugs for human use is lengthy, unpredictable and subject to many risks. We expect to continue incurring substantial expenses for the next several years as we continue to develop our clinical and preclinical drug candidates and programs. We are unable, with any certainty, to estimate either the costs or the timelines in which those expenses will be incurred. Our current development plans focus on the following programs: our lead respiratory program, PRS-060/AZD1402 and our other respiratory programs, our IO programs, currently comprised of cinrebafusp alfa as well as multiple additional proprietary and partnered programs, including PRS-344. These programs consume a large proportion of our current, as well as projected, resources.
Our research and development costs include costs that are directly attributable to the creation of certain of our Anticalin drug candidates and are comprised of:
 
internal recurring costs, such as personnel-related costs (salaries, employee benefits, equity compensation and other costs), materials and supplies, facilities and maintenance costs attributable to research and development functions; and

fees paid to external parties who provide us with contract services, such as preclinical testing, manufacturing and related testing and clinical trial activities.
General and Administrative Expenses
General and administrative expenses consist primarily of salaries, employee benefits, equity compensation and other personnel-related costs associated with executive, administrative and other support staff. Other significant general and administrative expenses include the costs associated with professional fees for accounting, auditing, insurance costs, consulting and legal services along with facility and maintenance costs attributable to general and administrative functions.

Results of Operations
Comparison of the three and six months ended June 30, 2021 and 2020
31


The following table sets forth our revenues and operating expenses (in thousands):
Three Months Ended June 30,Six Months Ended June 30,
2021202020212020
Revenues$3,285 $11,246 $18,918 $24,507 
Research and development expenses15,800 11,333 32,362 24,091 
General and administrative expenses4,246 4,568 8,376 8,927 
Total operating expenses20,046 15,901 40,738 33,018 
Other (expense) income
Interest income129 448 
Grant income796 — 796 — 
Other (expense) income, net464 (424)1,348 (484)
Net loss$(15,498)$(4,950)$(19,670)$(8,547)
Revenues
The following table provides a comparison of revenues for the three months ended June 30, 2021 and 2020 (in thousands):
 Three Months Ended June 30, 
 20212020Increase/(Decrease)
Customer revenue$2,540 $10,930 $(8,390)
Collaboration revenue745 316 429 
Total Revenue
$3,285 $11,246 (7,961)
 
The $8.4 million decrease in customer revenue in the three months ended June 30, 2021 compared to the three months ended June 30, 2020 relates to higher Seattle Genetics revenue recorded in the prior year upon the execution of a contractual amendment (approximately $3.5 million impact) as well as the achievement of a $5.0 million milestone on the first collaboration program.
The $0.4 million increase in collaboration revenues in the three months ended June 30, 2021 compared to the three months ended June 30, 2020 is due to higher amounts of cost-sharing revenue driven by increased manufacturing costs incurred with respect to our collaboration agreement with Servier.
The following table provides a comparison of revenues for the six months ended June 30, 2021 and 2020 (in thousands):
 Six Months Ended June 30, 
 20212020Increase/(Decrease)
Customer revenue$17,406 $19,815 $(2,409)
Collaboration revenue1,512 4,692 (3,180)
Total Revenue
$18,918 $24,507 (5,589)
 
The $2.4 million decrease in customer revenue in the six months ended June 30, 2021 compared to the six months ended June 30, 2020 relates to the phase 2a milestone ($13.0 million) recognized for PRS-060 under the AstraZeneca collaboration and higher pass through costs for Servier in the current year offset by higher Seattle Genetics revenue recorded upon the execution of a contractual amendment (approximately $3.5 million) and achievement of a $5.0 million milestone on the first collaboration program. There was also higher revenue recognized in the prior year related to a preclinical stage product that is not being pursued under the Servier collaboration.
The $3.2 million decrease in collaboration revenues in the six months ended June 30, 2021 compared to the six months ended June 30, 2020 relates to revenue recognized in the prior year on a preclinical stage product that Servier declined
32


to pursue further, offset slightly by higher Servier cost-sharing revenue generated from higher levels of manufacturing activities in the current year.
Research and Development Expenses
The following table provides a comparison of the research and development expenses for the three months ended June 30, 2021 and 2020 (in thousands):
 Three Months Ended June 30, 
 20212020Increase/(Decrease)
Respiratory$4,605 $2,697 $1,908 
Immuno-oncology4,495 2,585 1,910 
Other R&D activities6,707 6,051 656 
Total
$15,807 $11,333 4,474 
 
The $1.9 million increase in our respiratory programs for the three months ended June 30, 2021 compared to the three months ended June 30, 2020 is due to higher preclinical work being performed for our partnered and proprietary respiratory programs, with the majority of the increase attributed to PRS-220.
The $1.9 million increase in our IO programs for the three months ended June 30, 2021 compared to the three months ended June 30, 2020 is due primarily to an increase in manufacturing costs for cinrebafusp alfa and PRS-344 in addition to increased license fees due to the Boston Pharmaceuticals agreement.
The $0.7 million increase in other research and development activities expenses for the three months ended June 30, 2021 compared to the three months ended June 30, 2020 is due primarily to higher personnel and recruiting costs due to higher headcount, license fees, and external consulting expense offset slightly by lower facility costs due to the move to the new R&D facility in Hallbergmoos, Germany in the prior year.
The following table provides a comparison of the research and development expenses for the six months ended June 30, 2021 and 2020 (in thousands):
 Six Months Ended June 30, 
 20212020Increase/(Decrease)
Respiratory$8,633 $5,425 $3,208 
Immuno-oncology10,420 6,021 4,399 
Other R&D activities13,316 12,645 671 
Total
$32,369 $24,091 8,278 
 
The $3.2 million increase in our respiratory programs in the six months ended June 30, 2021 compared to the six months ended June 30, 2020 is due to higher preclinical work being performed on PRS-220, offset partially by lower clinical costs and manufacturing costs with respect to activities for PRS-060.
The $4.4 million increase in our IO programs in the six months ended June 30, 2021 compared to the six months ended June 30, 2020 is due primarily to an increase in manufacturing costs for cinrebafusp alfa.
The $0.7 million increase in other research and development activities expenses for the six months ended June 30, 2021 compared to the six months ended June 30, 2020 is due primarily to higher external consulting, personnel and recruiting costs due to higher headcount and license fees, offset slightly by lower facility costs due to the move to the new R&D facility in Hallbergmoos, Germany in the prior year.
General and Administrative Expenses
33


General and administrative expenses were $4.2 million for the three months ended June 30, 2021 and $4.6 million for the three months ended June 30, 2020. The period-over-period decrease is due primarily to lower legal and project management costs along with higher one-time office and building equipment costs incurred related to the move to the new R&D facility in Hallbergmoos, Germany in the prior year.
General and administrative expenses were $8.4 million for the six months ended June 30, 2021 and $8.9 million for the six months ended June 30, 2020. The period-over-period decrease is due primarily to higher legal and project management costs along with higher one-time office and building equipment costs related to the move to the new R&D facility in Hallbergmoos, Germany in the prior year, offset slightly by higher insurance and higher rent and depreciation expenses for the new R&D facility in the current year.
Other Income (Expense)
Our other income (expense) was $1.3 million for the three months ended June 30, 2021 and $(0.3) million for the three months ended June 30, 2020. This period over period increase was due to grant income recorded on PRS-220 as well as foreign exchange realized gains on upfront payments related to new collaboration and other milestone payments received.
Our other income was $2.2 million for the six months ended June 30, 2021 and de minimis for the six months ended June 30, 2020. This period over period increase was due to grant income recorded on PRS-220 as well as foreign exchange realized gains on upfront payment related to new collaboration and other milestone payments received.
Liquidity and Capital Resources
We are subject to risks common to companies in the biotechnology industry, including but not limited to, the need for additional capital, risks of failure of preclinical studies and clinical trials, the need to obtain marketing approval and reimbursement for any drug product candidate that we may identify and develop, the need to successfully commercialize and gain market acceptance of our product candidates, dependence on key personnel, protection of proprietary technology, compliance with government regulations, development of technological innovations by competitors, reliance on third-party manufacturers and the ability to transition from pilot-scale production to large-scale manufacturing of products.
Through June 30, 2021, we have funded our operations primarily through private and public sales of equity, payments received under our license and collaboration agreements (including research and development services costs, upfront and milestone payments), government grants and loans.
As of June 30, 2021, we had a total of $119.1 million in cash, cash equivalents and investments. We have incurred losses in every period since inception including the three and six months ended June 30, 2021 and 2020, respectively, and have a total accumulated deficit of $231.1 million as of June 30, 2021.
We have several research and development programs underway in varying stages of development, and we expect they will continue to require increasing amounts of cash for development, conducting clinical trials and testing and manufacturing of product material. We expect cash necessary to fund operations will increase significantly over the next several years as we continue to conduct these activities necessary to pursue governmental regulatory approval of clinical-stage programs and our other product candidates.
The following table provides a summary of operating, investing and financing cash flows (in thousands):
Six Months Ended June 30,
20212020
Net cash provided by (used in) operating activities$17,768 $(24,909)
Net cash (used in) provided by investing activities(257)6,010 
Net cash provided by financing activities32,310 416 

Net cash provided by operating activities for the six months ended June 30, 2021 and 2020 was $17.8 million and $24.9 million, respectively. Cash provided in 2021 is impacted by higher deferred revenue, primarily driven by the new collaboration agreements with Boston Pharmaceuticals and Genentech and higher accounts payable and accrued expenses, offset partially by higher accounts receivables and prepaid expenses. This compares to the impact of lower accounts payable, accrued expenses and deferred revenue, primarily driven by revenue recognized for the discontinued Servier programs and the satisfaction of a performance obligation under the Seattle Genetics agreements, for the six months ended June 30, 2020.
34


The change in net cash used in investing activities for the six months ended June 30, 2021 compared to net cash provided by investing activities for the same period in 2020 is mainly attributable to a significantly lower amount of purchases of property and equipment, as the purchases in prior year period primarily related to our move to a new R&D facility. Additionally, investing activities for the six months ended June 30, 2020 included the impact of net investments changes (lower purchases and increased maturities resulting in an overall decrease in investments) for which there is no activity in the comparable current year period.
Financing activities for the six months ended June 30, 2021 and 2020 provided cash of $32.3 million and $0.4 million, respectively. The change is driven by equity investments from both Seagen and AstraZeneca (see Note 3), sales under our ATM program, and proceeds received from warrant and option exercises. There was limited option exercise activity for the same period in 2020.
In August 2019, we entered into a sales agreement pursuant to which we may offer and sell shares of our common stock, from time to time, up to an aggregate gross sales proceeds of $50.0 million through our ATM Program, under a shelf registration statement on Form S-3 (File No. 333-226725). Through June 30, 2021, we have sold $22.7 million of common shares under the ATM Program of which 3.0 million shares were sold during the quarter ending June 30, 2021 at an average stock price of $4.24.
Our future success is dependent on our ability to identify and develop our product candidates, expand our corporate infrastructure and, ultimately, upon our ability to attain profitable operations. We have devoted substantially all of our financial resources and efforts to research and development and general and administrative expenses to support such research and development. We have several research and development programs underway in varying stages of development, and we expect that these programs will continue to require increasing amounts of cash for development, conducting clinical trials and testing and manufacturing of product material. Cash necessary to fund operations will increase significantly over the next several years as we continue to conduct these activities necessary to pursue governmental regulatory approval of clinical-stage programs and other product candidates.
Any requirements for additional capital will depend on many factors, including the following:
 
the scope, rate of progress, results and cost of our clinical studies, preclinical testing and other related activities;
the cost of manufacturing clinical supplies, and establishing commercial supplies, of our drug candidates and any products that we may develop;
the number and characteristics of drug candidates that we pursue;
the cost, timing and outcomes of regulatory approvals;
the cost and timing of establishing sales, marketing and distribution capabilities;
the terms and timing of any collaborative, licensing and other arrangements that we may establish;
the timing, receipt and amount of sales, profit sharing or royalties, if any, from our potential products;
the cost of preparing, filing, prosecuting, defending and enforcing any patent claims and other intellectual property rights;
the extent to which we acquire or invest in businesses, products or technologies, although we currently have no commitments or agreements relating to any of these types of transactions; and
the effects of the COVID-19 pandemic and the cost and timing of actions taken to contain it.
In addition, any unfavorable development or delay in the progress of our core clinical-stage programs including cinrebafusp alfa and PRS-060/AZD1402 could have a material adverse impact on our ability to raise additional capital.
We plan to raise additional capital to fulfill our operating and capital requirements through public or private equity financings, utilization of our current ATM Program, strategic collaborations, licensing arrangements and/or the achievement of milestones under our collaborative agreements. The funding requirements of our operating plans, however, are based on estimates that are subject to risks and uncertainties and may change as a result of many factors currently unknown. Although we continue to pursue these funding plans, there is no assurance that we will be successful in obtaining sufficient funding on terms acceptable to us to fund continuing operations, if at all. Until such time as we can generate substantial product revenues, if ever, we expect to finance our cash needs through a combination of equity offerings, debt financings, strategic partnerships, licensing arrangements and government grants. The terms of any future financing may adversely affect the holdings or the rights of our existing stockholders.
35


We believe that our currently available funds will be sufficient to fund our operations through at least the next 12 months from the issuance of this Quarterly Report on Form 10-Q. Our belief with respect to our ability to fund operations is based on estimates that are subject to risks and uncertainties. If actual results are different from our estimates, we may need to seek additional funding. If we are unable to obtain additional funding on acceptable terms when needed, we may be required to defer or limit some or all of our research, development and/or clinical projects.
Off-Balance Sheet Arrangements
We do not have any off-balance sheet arrangements, as defined under applicable SEC rules.
Critical Accounting Policies and Estimates
Refer to Part II, Item 7, “Critical Accounting Policies and Estimates” of our Annual Report on Form 10-K for the fiscal year ended on December 31, 2020 for a discussion of our critical accounting policies and estimates.
This discussion and analysis of our financial condition and results of operations is based on our financial statements, which we have prepared in accordance with U.S. GAAP. We believe that several accounting policies are important to understanding our historical and future performance. We refer to these policies as critical because these specific areas generally require us to make judgments and estimates about matters that are uncertain at the time we make the estimate, and different estimates—which also would have been reasonable—could have been used. On an ongoing basis, we evaluate our estimates and judgments, including those described in greater detail below. We base our estimates on historical experience and other market-specific or other relevant assumptions that we believe to be reasonable under the circumstances, the results of which form the basis for making judgments about the carrying value of assets and liabilities that are not readily apparent from other sources. Actual results may differ from these estimates under different assumptions or conditions.
We believe that our most critical accounting policies are those relating to revenue recognition, contingencies, research and development expense and income taxes, and there have not been significant changes to our accounting policies discussed in the Annual Report on Form 10-K for the fiscal year ended on December 31, 2020. 
Recently Issued Accounting Pronouncements
We review new accounting standards to determine the expected financial impact, if any, that the adoption of each standard will have. For the recently issued accounting standards that we believe may have an impact on our consolidated financial statements, see “Note 2—Summary of Significant Accounting Policies” in our consolidated financial statements.
Smaller Reporting Company Status
Currently, we qualify as a smaller reporting company.
As a smaller reporting company, we are eligible for, and have taken advantage of certain exemptions from various reporting requirements that are not available to public reporting companies that do not qualify for this classification, including, but not limited to:

An opportunity for reduced disclosure obligations regarding executive compensation in its periodic and annual reports, including without limitation exemption from the requirement to provide a compensation discussion and analysis describing compensation practices and procedures.

An opportunity for reduced financial statement disclosure in registration statements, which must include two years of audited financial statements rather than the three years of audited financial statements that are required for other public reporting companies.

An opportunity for reduced audit and other compliance expenses as we are not subject to the requirement to obtain an auditor’s report on internal control over financial reporting under Section 404(b) of the Sarbanes-Oxley Act of 2002.

An opportunity to continue utilizing the non-accelerated filer time-line requirements, which became applicable to us at the time of filing of our annual report for the year ending December 31, 2020.
For as long as we continue to be a smaller reporting company, we expect that we will take advantage of both the reduced internal control audit requirements and the disclosure obligations available to us as a result of this classification.

Item 3. Quantitative and Qualitative Disclosures About Market Risk
36



We are a smaller reporting company as defined by Rule 12b-2 of the Exchange Act and are not required to provide the information required under this item.

Item 4. Controls and Procedures.

Evaluation of Disclosure Controls and Procedures

Our management, with the participation of our principal executive officer and principal financial officer, have evaluated the effectiveness of our disclosure controls and procedures as of the end of the period covered by this Quarterly Report on Form 10-Q.  The term “disclosure controls and procedures,” as defined in Rules 13a-15(e) and 15d-15(e) under the Exchange Act, means controls and other procedures of a company that are designed to ensure that information required to be disclosed by us in the reports that we file or submit under the Exchange Act is recorded, processed, summarized and reported, within the time periods specified in the SEC’s rules and forms, and such information is accumulated and communicated to our management, including our principal executive and principal financial officers, or persons performing similar functions, as appropriate to allow timely decisions regarding required disclosure.  Our principal executive officer and principal financial officer have concluded that, based on such evaluation, our disclosure controls and procedures were effective as of June 30, 2021.
Changes in Internal Control over Financial Reporting

There have been no changes in our internal control over financial reporting identified in connection with the evaluation of such internal control required by Rules 13a-15(d) and 15d-15(d) under the Exchange Act that occurred during the quarter ended June 30, 2021 that have materially affected, or are reasonably likely to materially affect, our internal control over financial reporting.

37


PART II - OTHER INFORMATION
 
Item 1. Legal Proceedings.
As of the date of this Quarterly Report on Form 10-Q, we are not party to and our property is not subject to any material pending legal proceedings. However, from time to time, we may become involved in legal proceedings or subject to claims that arise in the ordinary course of our business activities. Regardless of the outcome, such legal proceedings or claims could have an adverse impact on us because of defense and settlement costs, diversion of management resources and other factors.

Item 1A. Risk Factors.
Please refer to the complete Item 1A of the Company’s Annual Report on Form 10-K for the fiscal year ended December 31, 2020, filed with the SEC on March 31, 2021 for risks and uncertainties facing the Company that may have a material adverse effect on the Company’s business prospects, financial condition and results of operations. There have been no material changes in the risk factors described in our Annual Report on Form 10-K for the fiscal year ended December 31, 2020.

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

None.

Item 3. Defaults Upon Senior Securities.

None.

Item 4. Mine Safety Disclosures.

Not applicable.

Item 5. Other Information.
None.

Item 6. Exhibits.
Exhibit
Number
Exhibit DescriptionIncorporated by
Reference herein
from Form or
Schedule
Filing DateSEC File /
Registration
Number
Certificate of Designation of Series E Convertible Preferred Stock of Pieris Pharmaceuticals, Inc.Form 8-K (Exhibit 3.1)May 21, 2021001-37471
Exclusive Product License Agreement, dated April 24, 2021, by and among Pieris Pharmaceuticals, Inc., Pieris Pharmaceuticals GmbH and BP Asset XII, Inc.
*
Exchange Agreement by and among Pieris Pharmaceuticals, Inc. and Biotechnology Value Fund, L.P., Biotechnology Value Fund II, L.P., Biotechnology Value Trading Fund OS, L.P., and MSI BVF SPV, L.L.C., dated as of May 20, 2021.
Form 8-K (Exhibit 10.1)May 21, 2021001-37471
Research Collaboration and License Agreement, dated May 19, 2021, by and among Pieris Pharmaceuticals, Inc., Pieris Pharmaceuticals GmbH and Genentech, Inc.
*
38


Exhibit
Number
Exhibit DescriptionIncorporated by
Reference herein
from Form or
Schedule
Filing DateSEC File /
Registration
Number
Pieris Pharmaceuticals, Inc. 2020 Employee, Director and Consultant Equity Incentive Plan, as Amended.
Form 8-K (Exhibit 10.1)June 29, 2021001-37471
  Certification of Principal Executive Officer Pursuant to Rules 12a-14(a) and 15d-14(a) under the Securities Exchange Act of 1934, as Adopted Pursuant to Section 302 of the Sarbanes-Oxley Act of 2002.*   
  Certification of Principal Financial Officer and Principal Accounting Officer Pursuant to Rules 12a-14(a) and 15d-14(a) under the Securities Exchange Act of 1934, as Adopted Pursuant to Section 302 of the Sarbanes-Oxley Act of 2002.*   
  Certification of Principal Executive Officer Pursuant to 18 U.S.C Section 1350, as Adopted Pursuant to Section 906 of the Sarbanes-Oxley Act of 2002.**   
  Certification of Principal Financial Officer and Principal Accounting Officer Pursuant to 18 U.S.C Section 1350, as Adopted Pursuant to Section 906 of the Sarbanes-Oxley Act of 2002.**   
101.INS  Inline XBRL Instance Document (the instance document does not appear in the Interactive Data File because its XBRL tags are embedded within the Inline XBRL document)   
101.SCH  Inline XBRL Taxonomy Extension Schema Document*   
101.CAL  Inline XBRL Taxonomy Extension Calculation Linkbase Document*   
101.DEF  Inline XBRL Taxonomy Extension Definition Linkbase Document*   
101.LAB  Inline XBRL Taxonomy Extension Label Linkbase Document*   
101.PRE  Inline XBRL Taxonomy Extension Presentation Linkbase Document*   
104Cover Page Interactive Data File (formatted as inline XBRL and contained in Exhibit 101)*
*Filed herewith.
**The certifications furnished in Exhibit 32.1 and Exhibit 32.2 hereto are deemed to accompany this Quarterly Report and will not be deemed “filed” for purposes of Section 18 of the Securities Exchange Act of 1934, as amended, except to the extent that the registrant specifically incorporates it by reference.
39


Exhibit
Number
Exhibit DescriptionIncorporated by
Reference herein
from Form or
Schedule
Filing DateSEC File /
Registration
Number
±Certain confidential portions of this Exhibit were omitted by means of marking such portions with brackets (“[***]”) because the identified confidential portions (i) are not material and (ii) would be competitively harmful if publicly disclosed.

40


SIGNATURES
Pursuant to the requirements of the Securities Exchange Act of 1934, the registrant has duly caused this Quarterly Report on Form 10-Q to be signed on its behalf by the undersigned thereunto duly authorized.
PIERIS PHARMACEUTICALS, INC.
August 5, 2021By: /s/ Stephen S. Yoder
 Stephen S. Yoder
 Chief Executive Officer and President
(Principal Executive Officer)
August 5, 2021By:/s/ Thomas Bures
Thomas Bures
Vice President, Finance and Treasurer
(Principal Financial Officer and Principal Accounting Officer)

41