SANDPIPER: A Study of Taselisib + Fulvestrant Versus Placebo + Fulvestrant in Participants With Advanced or Metastatic Breast Cancer Who Have Disease Recurrence or Progression During or After Aromatase Inhibitor Therapy

Sponsor
Hoffmann-La Roche (Industry)
Overall Status
Terminated
CT.gov ID
NCT02340221
Collaborator
(none)
631
157
2
74.7
4
0.1

Study Details

Study Description

Brief Summary

This international, multicenter, randomized, double-blinded, placebo-controlled study is designed to compare the efficacy and safety of taselisib + fulvestrant with that of placebo + fulvestrant in postmenopausal women with estrogen receptor (ER)-positive, human epidermal growth factor receptor-2 (HER2)-negative, oncogene that encodes for phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3CA)-mutant, unresectable, locally advanced or metastatic breast cancer after recurrence or progression during or after an aromatase inhibitor (AI) therapy. There will be a 2:1 randomization to the taselisib arm versus the placebo arm. Enrollment will be enriched for participants with PIK3CA mutant tumors via central testing. The anticipated duration of the study is approximately 3.5 years.

Condition or Disease Intervention/Treatment Phase
Phase 3

Study Design

Study Type:
Interventional
Actual Enrollment :
631 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
Double (Participant, Investigator)
Primary Purpose:
Treatment
Official Title:
A Phase III, Double-Blind, Placebo-Controlled, Randomized Study of Taselisib Plus Fulvestrant Versus Placebo Plus Fulvestrant in Postmenopausal Women With Estrogen Receptor-Positive and HER2-Negative Locally Advanced or Metastatic Breast Cancer Who Have Disease Recurrence or Progression During or After Aromatase Inhibitor Therapy
Actual Study Start Date :
Apr 9, 2015
Actual Primary Completion Date :
Jun 29, 2021
Actual Study Completion Date :
Jun 29, 2021

Arms and Interventions

Arm Intervention/Treatment
Experimental: Taselisib + Fulvestrant

Participants received taselisib 4 milligrams (mg) taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.

Drug: Taselisib
Taselisib 4 mg was administered as two tablets of 2 mg each as per the schedule specified in the respective arm.
Other Names:
  • GDC-0032, RO5537381
  • Drug: Fulvestrant
    Fulvestrant 500 mg was administered as two IM injections of 250 mg each as per the schedule specified in the respective arms.
    Other Names:
  • Faslodex
  • Placebo Comparator: Placebo + Fulvestrant

    Participants received placebo taken orally once daily (QD) beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by intramuscular (IM) injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.

    Drug: Placebo
    Placebo matching to taselisib was administered as per the schedule specified in the respective arm.

    Drug: Fulvestrant
    Fulvestrant 500 mg was administered as two IM injections of 250 mg each as per the schedule specified in the respective arms.
    Other Names:
  • Faslodex
  • Outcome Measures

    Primary Outcome Measures

    1. Progression-Free Survival (PFS) as Assessed by Investigator Using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) at Primary Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)]

      PFS was defined as the time from randomization to disease progression as determined by the investigator with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 millimeters (mm). For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    2. PFS as Assessed by Investigator Using RECIST v1.1 at Final Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)]

      PFS was defined as the time from randomization to disease progression as determined by the investigator with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    Secondary Outcome Measures

    1. Percentage of Participants With Objective Response (Partial Response [PR] Plus Complete Response [CR]), as Assessed Using RECIST v.1.1 at Primary Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)]

      PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions).

    2. Percentage of Participants With Objective Response (PR Plus CR), as Assessed Using RECIST v.1.1 at Final Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)]

      PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions).

    3. Overall Survival (OS) at Primary Analysis [From randomization up to death from any cause (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)]

      OS was defined as the time from the date of randomization to the date of death due to any cause.

    4. OS at Final Analysis [From randomization up to death from any cause (up to approximately 6.2 years)]

      OS was defined as the time from the date of randomization to the date of death due to any cause.

    5. Percentage of Participants With Clinical Benefit, as Assessed According to RECIST v1.1 at Primary Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)]

      Clinical benefit was defined as objective response (PR+CR), or no disease progression lasting for more than or equal to (>/=) 24 weeks since randomization. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    6. Percentage of Participants With Clinical Benefit, as Assessed According to RECIST v1.1 at Final Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)]

      Clinical benefit was defined as objective response (PR+CR), or no disease progression lasting for more than or equal to (>/=) 24 weeks since randomization. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    7. Duration of Objective Response, as Assessed by Investigator Using RECIST v1.1 at Primary Analysis [Time from the first occurrence of a documented objective response to the time of the first documented disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)]

      Duration of objective response: the time from the first tumor assessment that supported the participant's objective response (CR or PR, whichever was first recorded) to first documented disease progression or death due to any cause, whichever occurred first. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. Disease progression: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    8. Duration of Objective Response, as Assessed by Investigator Using RECIST v1.1 at Final Analysis [Time from the first occurrence of a documented objective response to the time of the first documented disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)]

      Duration of objective response: the time from the first tumor assessment that supported the participant's objective response (CR or PR, whichever was first recorded) to first documented disease progression or death due to any cause, whichever occurred first. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. Disease progression: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    9. PFS as Assessed by Blinded Independent Central Review (BICR) Using RECIST v1.1 at Primary Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)]

      PFS was defined as the time from randomization to disease progression as determined by BICR with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    10. PFS as Assessed by BICR Using RECIST v1.1 at Final Analysis [From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)]

      PFS was defined as the time from randomization to disease progression as determined by BICR with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.

    11. Percentage of Participants With Adverse Events at Primary Analysis [From randomization up to the 15 Oct 2017 data cutoff, approximately 2.5 years.]

      An adverse event was any untoward medical occurrence in a clinical investigation participant administered a pharmaceutical product, regardless of causal attribution.

    12. Percentage of Participants With Adverse Events at Final Analysis [From randomization up to approximately 6.2 years]

      An adverse event was any untoward medical occurrence in a clinical investigation participant administered a pharmaceutical product, regardless of causal attribution.

    13. Maximum Observed Plasma Concentration (Cmax) of Taselisib [1 to 4 hours (hrs) post-dose on Cycle (C) 1, Day (D) 1; 0 to 3 hrs pre-dose and 2 to 6 hrs post dose on Cycle 2, Day 1 (each cycle=28 days)]

    14. Minimum Observed Plasma Concentration (Cmin) of Taselisib [1 to 4 hrs post-dose on Cycle 1, Day 1; 0 to 3 hrs pre-dose and 2 to 6 hrs post dose on Cycle 2, Day 1; 0 to 3 hrs pre-dose on Cycle 6, Day 1 (each cycle=28 days)]

    15. Change From Baseline in European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Core 30 (QLQ-C30) Score [Baseline, C2D1 up to C7D1 (each cycle=28 days)]

      The EORTC QLQ-C30 consists of 30 questions that comprise aspects of participant's functioning assessment (physical, emotional, role, cognitive, and social); symptom scales (fatigue; nausea, vomiting, and pain; the global health/quality of life [QoL]); and single items (dyspnoea, insomnia, appetite loss, constipation, diarrhoea, and financial difficulties), within a recall period of "the past week." Most questions used a 4-point scale (1=Not at all to 4=Very much; two questions used a 7-point scale (1=Very poor to 7=Excellent). Scores were averaged and transformed to a 0-100 scale; a higher score for Global Qol/functional scales=better level of functioning; a higher score for symptom scale=greater degree of symptoms.

    16. Change From Baseline in Modified EORTC Quality of Life Questionnaire Breast Cancer Module 23 (QLQ-BR23) Score [Baseline, C2D1 up to C7D1 (each cycle=28 days)]

      EORTC-QLQ-BR23 is a 23-item breast cancer-specific companion module to the EORTC-QLQ-C30 and consists of functional scales (body image, sexual enjoyment, sexual functioning, future perspective [FP]) and symptom scales (systemic side effects [SE], upset by hair loss, arm symptoms, breast symptoms). Questions used a 4-point scale (1=not at all, 2=a little, 3=quite a bit, 4=very much). Scores were averaged and transformed to a 0-100 scale. Higher scores for the functional scales indicated a higher/better level of functioning/healthy functioning. Higher scores for the symptom scales indicated worse symptoms.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years and Older
    Sexes Eligible for Study:
    Female
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    • Postmenopausal women with histologically or cytologically confirmed locally advanced or metastatic estrogen receptor (ER) positive breast cancer

    • Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1

    • Participants for whom endocrine therapy (example [e.g.], fulvestrant) is recommended and treatment with cytotoxic chemotherapy is not indicated at time of entry into the study

    • Radiologic/objective evidence of recurrence or progression to the most recent systemic therapy for breast cancer

    • Radiologic/objective evidence of breast cancer recurrence or progression while on or within 12 months of the end of adjuvant treatment with an aromatase inhibitor (AI), or progression while on or within 1 month of the end of prior AI treatment for locally advanced or metastatic breast cancer

    • Measurable disease via Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) or non-measurable, evaluable disease with at least one evaluable bone lesion via RECIST v1.1

    • Consent to provide a formalin-fixed, paraffin-embedded (FFPE) tumor tissue block (preferred) or a minimum of 20 (25 preferred) freshly cut unstained tumor slides from the most recently collected, available tumor tissue for oncogene that encodes for phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3CA)-mutation testing

    • A valid cobas PIK3CA mutation result by central testing is required

    • Adequate hematologic and end-organ function within 28 days prior to treatment initiation

    Exclusion Criteria:
    • Human epidermal growth factor receptor 2 (HER2)-positive disease by local laboratory testing (immunohistochemistry 3 positive [IHC 3+] staining or in situ hybridization positive)

    • Prior treatment with fulvestrant

    • Prior treatment with a phosphatidylinositol 3-kinase (PI3K) inhibitor, mammalian target of rapamycin (mTOR) inhibitor (e.g. everolimus), or protein kinase B (AKT) inhibitor

    • Prior anti-cancer therapy within 2 weeks prior to Day 1 of Cycle 1

    • Prior radiation therapy within 2 weeks prior to Day 1 of Cycle 1

    • All acute treatment-related toxicity must have resolved to Grade less than or equal to (</=) 1 or be deemed stable by the Investigator

    • Prior treatment with greater than (>) 1 cytotoxic chemotherapy regimen for metastatic breast cancer

    • Concurrent hormone replacement therapy

    • Known untreated or active central nervous system (CNS) metastases

    • Type 1 or Type 2 diabetes mellitus requiring anti-hyperglycemic medications

    • History of inflammatory bowel disease or active bowel inflammation

    • Clinically significant cardiac or pulmonary dysfunction

    • Clinically significant history of liver disease, including cirrhosis, current alcohol abuse, or current known active infection with human immunodeficiency virus (HIV), hepatitis B or C virus

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Arizona Oncology Tucson Arizona United States 85704
    2 Arizona Oncology Associates, P.C. Tucson Arizona United States 85710
    3 Georgia Cancer Specialists - Northside Atlanta Georgia United States 30341
    4 Northwest Georgia Oncology Centers, a Service of WellStar Cobb Hospital Marietta Georgia United States 30060
    5 Ingalls Hospital Harvey Illinois United States 60426
    6 Maryland Oncology Hematology Rochville Maryland United States 20850
    7 Dana Farber Can Ins Boston Massachusetts United States 02215
    8 Mercy Hospitals East Communities d/b/a Mercy Hospital St. Louis Saint Louis Missouri United States 63141
    9 MSKCC at Basking Ridge Basking Ridge New Jersey United States 07920
    10 John Theurer Cancer Ctr at Hackensack Univ Medical Ctr Hackensack New Jersey United States 07601
    11 Memorial Sloan-Kettering; Cancer Center Commack New York United States 11725
    12 Memorial Sloan Kettering Cancer Center; Memorial Sloan Kettering; at Westchester Harrison New York United States 10604
    13 Memorial Sloan Kettering Cancer Center New York New York United States 10065
    14 Memorial Sloan Kettering Nassau Uniondale New York United States 11553
    15 Oregon Health & Science University; Knight Cancer Institute, Community Hematology Oncology Beaverton Oregon United States 97006
    16 Pinnacle Health Harrisburg Pennsylvania United States 17110
    17 Liverpool Hospital; Cancer Therapy Centre Liverpool New South Wales Australia 2170
    18 Macquarie University Hospital Macquarie Park New South Wales Australia 2109
    19 Newcastle Mater Misericordiae Hospital; Oncology Waratah New South Wales Australia 2298
    20 Mater Hospital; Oncology Brisbane Queensland Australia 4101
    21 Austin Hospital; Medical Oncology Heidelberg Victoria Australia 3084
    22 Sunshine Hospital; Oncology Research St Albans Victoria Australia
    23 St John of God Murdoch Hospital; Oncology West Murdoch Western Australia Australia 6150
    24 Lkh-Univ. Klinikum Graz; Klinik Für Innere Medizin I Graz Austria 8036
    25 Tiroler Landeskrankenanstalten Ges.M.B.H.; Abt. Für Gynäkologie Innsbruck Austria 6020
    26 Ordensklinikum Linz Barmherzige Schwestern; Abt. fur Innere Medizin 1 Linz Austria 4010
    27 Medizinische Universität Wien; Univ.Klinik für Innere Medizin I Wien Austria 1090
    28 University Clinical Center of the Republic of Srpska Banja Luka Bosnia and Herzegovina 78000
    29 Clinic of Oncology, University Clinical Center Sarajevo Sarajevo Bosnia and Herzegovina 7100
    30 Complex Oncological Center - Plovdiv, EOOD Plovdiv Bulgaria 4004
    31 MHAT Nadezhda Sofia Bulgaria 1330
    32 SHATO - Sofia Sofia Bulgaria 1756
    33 SHATOD Dr. Marko Antonov Markov-Varna, EOOD Varna Bulgaria 9010
    34 Cross Cancer Institute Edmonton Alberta Canada T6G 1Z2
    35 British Columbia Cancer Agency (Bcca) - Vancouver Cancer Centre Vancouver British Columbia Canada V5Z 4E6
    36 Grand River Hospital Kitchener Ontario Canada N2G 1G3
    37 The Ottawa Hospital Cancer Centre; Oncology Ottawa Ontario Canada K1H 8L6
    38 Sunnybrook Health Science Centre Toronto Ontario Canada M4N 3M5
    39 McGill University; Sir Mortimer B Davis Jewish General Hospital; Oncology Montreal Quebec Canada H3T 1E2
    40 Hospital Du Saint-Sacrement Quebec City Quebec Canada G1S 4L8
    41 the First Hospital of Jilin University Changchun China 130021
    42 Jilin Cancer Hospital Changchun China 132013
    43 Jiangsu Cancer Hospital Nanjing City China 211100
    44 Fudan University Shanghai Cancer Center Shanghai City China 200120
    45 Zhejiang Cancer Hospital Zhejiang China 310022
    46 Clinica del Country Bogota Colombia 11001
    47 Oncomedica S.A. Monteria Colombia 230002
    48 University Hospital; Oncology and Radiotherapy Hradec Kralove Czechia 500 05
    49 Fakultni nemocnice Olomouc; Onkologicka klinika Olomouc Czechia 779 00
    50 Vseobecna fakultni nemocnice v Praze Praha 2 Czechia 128 08
    51 KYS Sadesairaala; Syopatautien poliklinikka Kuopio Finland 70210
    52 Turku Uni Central Hospital; Oncology Clinics Turku Finland 20520
    53 Centre Jean Perrin; Hopital De Semaine Clermont-Ferrand France 63011
    54 Centre Georges François Leclerc; Service Pharmacie, Bp 77980 Dijon France 21000
    55 Hopital Prive Drome Ardeche; Chir 2A 2B Guilherand Granges France 07500
    56 CHD Vendée La Roche Sur Yon France 85025
    57 Hopital Dupuytren; Oncologie Medicale Limoges France 87042
    58 Institut régional du Cancer Montpellier Montpellier France 34298
    59 Institut Curie; Oncologie Medicale Paris France 75231
    60 Ch Lyon Sud; Onco Secteur Jules Courmont Pierre Benite France 69495
    61 Pole de Cancerologie Prive Strasbourgeois Strasbourg France 67000
    62 Centre Alexis Vautrin; Oncologie Medicale Vandoeuvre-les-nancy France 54519
    63 Hochwaldkrankenhaus; Abt.Gynäkologie Geburtshilfe u.Senologie Bad Nauheim Germany 61231
    64 Praxisklinik Krebsheilkunde für Frauen / Brustzentrum (Dres. Kittel/Klare) Berlin Germany 10367
    65 Onkologische Schwerpunktpraxis Kurfürstendamm Berlin Germany 10707
    66 Universitätsklinikum Essen; Zentrum Für Frauenheilkunde Essen Germany 45122
    67 Kooperatives Mammazentrum Hamburg Krankenhaus Jerusalem Hamburg Germany 20357
    68 Klinikum der Universität München; Frauenklinik - Onkologie II München Germany 80337
    69 Klinikum Mutterhaus der Borromäerinnen, Innere Medizin I Trier Germany 54290
    70 Alexandras General Hospital of Athens; Oncology Department Athens Greece 115 28
    71 IASO General Hospital of Athens Athens Greece 155 62
    72 Univ General Hosp Heraklion; Medical Oncology Heraklion Greece 711 10
    73 University Hospital of Patras Medical Oncology Patras Greece 265 04
    74 Euromedical General Clinic of Thessaloniki; Oncology Department Thessaloniki Greece 546450
    75 Papageorgiou General Hospital; Medical Oncology Thessaloniki Greece 564 29
    76 Istituto Nazionale Tumori Irccs Fondazione g. PASCALE;U.O.C. Oncologia Medica Senologica Napoli Campania Italy 80131
    77 ARCISPEDALE S. MARIA NUOVA - REGGIO EMILIA; Struttura Semplice Coordinamento Breast Unit Integrata Reggio Emilia Emilia-Romagna Italy 42100
    78 A.O. Universitaria S. Maria Della Misericordia Di Udine; Oncologia Udine Friuli-Venezia Giulia Italy 33100
    79 IRCCS Istituto Nazionale Per La Ricerca Sul Cancro (IST); Oncologia Medica A Genova Liguria Italy 16132
    80 Istituto Europeo Di Oncologia Milano Lombardia Italy 20141
    81 Centro Catanese Di Oncologia; Oncologia Medica Catania Sicilia Italy 95126
    82 Azienda Ospedaliero-Universitaria Careggi; SOD Radioterapia Firenze Toscana Italy 50134
    83 Azienda USL 9 Grosseto; Dipartimento Politiche del Farmaco Grosseto Toscana Italy 58100
    84 Azienda usl 5 Di Pisa-Ospedale Di Pontedera;U.O. Oncologia Pontedera Toscana Italy 56025
    85 AZ. Usll12 Veneziana-Ospedale Dell'angelo;Oncologia Medica Mestre Veneto Italy 30174
    86 Inje university Haeundae Paik Hospital Busan Korea, Republic of 48108
    87 Chungbuk National University Hospital Cheongju-si Korea, Republic of 28644
    88 National Cancer Center Goyang-si Korea, Republic of 10408
    89 Korea University Anam Hospital Seoul Korea, Republic of 02841
    90 Seoul National University Hospital Seoul Korea, Republic of 03080
    91 Severance Hospital, Yonsei University Health System Seoul Korea, Republic of 03722
    92 Asan Medical Center Seoul Korea, Republic of 05505
    93 Samsung Medical Center Seoul Korea, Republic of 06351
    94 Ulsan University Hosiptal Ulsan Korea, Republic of 44033
    95 Iem-Fucam D.f. Mexico 04980
    96 Instituto Nacional De Cancerologia; Oncology; Tumores Mamarios Distrito Federal Mexico 14000
    97 Consultorio de Medicina Especializada; Dentro de Condominio San Francisco Mexico City Mexico 03100
    98 Hospital San Jose Del Tec. de Monterrey; Oncology Monterrey Mexico 64020
    99 Oaxaca Site Management Organization Oaxaca Mexico 68000
    100 Medisch Centrum Alkmaar Alkmaar Netherlands 1815 JD
    101 Ziekenhuis Rijnstate Arnhem Netherlands 6815 AD
    102 Instituto Nacional de Enfermedades Neoplasicas Lima Peru Lima 34
    103 Clinica Internacional, Sede San Borja; Unidad de Investigacion de Clínica Internacional Lima Peru Lima 41
    104 Oncocenter Peru S.A.C.; Oncosalud Lima Peru Lima 41
    105 Instituto Regional de Enfermedades Neoplasicas - IREN Norte Trujillo Peru 13014
    106 Bialostockie Centrum Onkologii; Oddzial Onkologii Klinicznej Bialystok Poland 15-027
    107 Centrum Onkologii w Bydgoszczy; Oddzial Kliniczny Onkologii Bydgoszcz Poland 85-796
    108 Uniwersyteckie Centrum Kliniczne, Klinika Onkologii i Radioterapii Gdansk Poland 80-214
    109 Przychodnia Lekarska KOMED, Roman Karaszewski Konin Poland 62-500
    110 Szpital Uniwersytecki w Krakowie, Oddział Kliniczny Kliniki Onkologii Kraków Poland 30-688
    111 Woj.Wielospecjalistyczne Centrum Onkologii i Traumatologii; Oddz.Hematologii Pododz.Chemioterapii Lodz Poland 93-513
    112 Centrum Onkologii Ziemi LUBELSKIEJ im. Sw Jana z Dukli, I oddz. Chemioterapii Lublin Poland 20-090
    113 Zachodniopomorskie Centrum Onkologii, Osrodek Innowacyjnosci, Rozwoju i Badan Klinicznych Szczecin Poland 71-730
    114 Centrum Onkologii - Instytut im. Marii Skłodowskiej-Curie Klinika Nowotworów Piersi i Chirurgii Warszawa Poland 02-781
    115 Wojewodzki Szpital Specjalistyczny; Osrodek Badawczo-Rozwojowy, Oddzial Chemioterapii Wroclaw Poland 51-124
    116 Hospital Garcia de Orta; Servico de Oncologia Medica Almada Portugal 2801-951
    117 IPO de Lisboa; Servico de Oncologia Medica Lisboa Portugal 1099-023
    118 Hospital da Luz; Departamento de Oncologia Medica Lisboa Portugal 1500-650
    119 Hospital de Santa Maria; Servico de Oncologia Medica Lisboa Portugal 1649-035
    120 IPO do Porto; Servico de Oncologia Medica Porto Portugal 4200-072
    121 Institut of Oncology Al. Trestioreanu Bucharest; Oncology Department Bucuresti Romania 022328
    122 Prof. Dr. I. Chiricuta Institute of Oncology Cluj Napoca Romania 400015
    123 Oncology Center Sf. Nectarie Craiova Romania 200347
    124 Euroclinic Center of Oncology SRL Iasi Romania 700106
    125 Moscow City Oncology Hospital #62 Moscovskaya Oblast Moskovskaja Oblast Russian Federation 143423
    126 Regional Clinical Oncology Dispensary; Surgery Dept, Thoracic Arkhangelsk Russian Federation 163045
    127 Ivanovo Regional Oncology Dispensary Ivanovo Russian Federation 153040
    128 Clinical Oncology Dispensary of Ministry of Health of Tatarstan Kazan Russian Federation 420029
    129 State Inst. Of Healthcare Orenburg Regional Clinical Oncology Dis Orenburg Russian Federation 460021
    130 S-Pb clinical scientific practical center of specialized kinds of medical care (oncological) Saint-Petersburg Russian Federation 197758
    131 Institute for Onc/Rad Serbia Belgrade Serbia 11000
    132 Complejo Hospitalario Universitario de Santiago (CHUS) ; Servicio de Oncologia Santiago de Compostela LA Coruña Spain 15706
    133 Hospital Universitario Puerta de Hierro; Servicio de Oncologia Majadahonda Madrid Spain 28222
    134 Hospital Universitario de Canarias;servicio de Oncologia La Laguna Tenerife Spain 38320
    135 Hospital de Cruces; Servicio de Oncologia Bilbao Vizcaya Spain 48903
    136 Hospital Universitari Vall d'Hebron Barcelona Spain 08035
    137 Hospital Clínic i Provincial; Servicio de Hematología y Oncología Barcelona Spain 08036
    138 Centro Oncologico MD Anderson Internacional; Servicio de Oncologia Madrid Spain 28033
    139 Hospital Ramon y Cajal; Servicio de Oncologia Madrid Spain 28034
    140 HOSPITAL DE MADRID NORTE SANCHINARRO- CENTRO INTEGRAL ONCOLOGICO CLARA CAMPAL; Servicio de Oncologia Madrid Spain 28050
    141 Fundación IVO Valencia Spain 46980
    142 Hospital Universitario Miguel Servet; Servicio Oncologia Zaragoza Spain 50009
    143 Uni Hospital Linkoeping; Dept. of Oncology Linköping Sweden 58185
    144 Sodersjukhuset; Onkologkliniken Stockholm Sweden 118 83
    145 Akademiska sjukhuset, Onkologkliniken Uppsala Sweden 751 85
    146 Koo Foundation Sun Yat-Sen Cancer Center; Hemato-Oncology Taipei City Taiwan 11259
    147 VETERANS GENERAL HOSPITAL; Department of General Surgery Taipei Taiwan 00112
    148 National Taiwan Uni Hospital; General Surgery Taipei Taiwan 100
    149 Mackay Memorial Hospital; Dept of Surgery Taipei Taiwan 104
    150 Department of Surgery, King Chulalongkorn Memorial Hospital Bangkok Thailand 10330
    151 Ramathibodi Hospital; Dept of Med.-Div. of Med. Onc Bangkok Thailand 10400
    152 Maharaj Nakorn Chiang Mai Hospital; Department of Surgery/Head Neck and Breast Unit; Clinical Trial Chiang Mai Thailand 50200
    153 Baskent University Adana Dr. Turgut Noyan Practice and Research Hospital; Medical Oncology Adana Turkey 01230
    154 Trakya University Medical Faculty Research And Practice Hospital Medical Oncology Department Edirne Turkey 22770
    155 Istanbul Uni Cerrahpasa Medical Faculty Hospital; Medical Oncology Istanbul Turkey 34300
    156 Ege Uni Medical Faculty; Oncology Dept Izmir Turkey 35100
    157 Hacettepe Uni Medical Faculty Hospital; Oncology Dept Sihhiye/Ankara Turkey 06230

    Sponsors and Collaborators

    • Hoffmann-La Roche

    Investigators

    • Study Director: Clinical Trials, Hoffmann-La Roche

    Study Documents (Full-Text)

    More Information

    Publications

    None provided.
    Responsible Party:
    Hoffmann-La Roche
    ClinicalTrials.gov Identifier:
    NCT02340221
    Other Study ID Numbers:
    • GO29058
    • 2014-003185-25
    First Posted:
    Jan 16, 2015
    Last Update Posted:
    Jul 12, 2022
    Last Verified:
    Jul 1, 2022
    Studies a U.S. FDA-regulated Drug Product:
    Yes
    Studies a U.S. FDA-regulated Device Product:
    No
    Additional relevant MeSH terms:

    Study Results

    Participant Flow

    Recruitment Details This study was conducted at 155 centers in 28 countries.
    Pre-assignment Detail The study enrolled postmenopausal women with estrogen receptor-positive and human epidermal receptor 2 (HER2)-negative locally advanced or metastatic breast cancer who had disease recurrence or progression during or after aromatase inhibitor therapy. Randomization was stratified by three factors: 1) visceral versus non-visceral disease, 2) sensitivity versus non-sensitivity to most recent endocrine therapy, and 3) geographical region.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Period Title: Overall Study
    STARTED 214 417
    COMPLETED 0 0
    NOT COMPLETED 214 417

    Baseline Characteristics

    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant Total
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Total of all reporting groups
    Overall Participants 214 417 631
    Age (years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [years]
    60.7
    (10.0)
    60.1
    (9.9)
    60.3
    (9.9)
    Sex: Female, Male (Count of Participants)
    Female
    214
    100%
    417
    100%
    631
    100%
    Male
    0
    0%
    0
    0%
    0
    0%
    Race/Ethnicity, Customized (Count of Participants)
    Not Hispanic or Latino
    166
    77.6%
    334
    80.1%
    500
    79.2%
    Hispanic or Latino
    26
    12.1%
    48
    11.5%
    74
    11.7%
    Not Stated
    15
    7%
    15
    3.6%
    30
    4.8%
    Unknown
    7
    3.3%
    20
    4.8%
    27
    4.3%
    Race/Ethnicity, Customized (Count of Participants)
    White
    147
    68.7%
    285
    68.3%
    432
    68.5%
    Asian
    38
    17.8%
    72
    17.3%
    110
    17.4%
    Unknown
    14
    6.5%
    30
    7.2%
    44
    7%
    American Indian or Alaska Native
    13
    6.1%
    24
    5.8%
    37
    5.9%
    Black or African American
    2
    0.9%
    2
    0.5%
    4
    0.6%
    Native Hawaiian or Other Pacific Islander
    0
    0%
    3
    0.7%
    3
    0.5%
    Multiple
    0
    0%
    1
    0.2%
    1
    0.2%

    Outcome Measures

    1. Primary Outcome
    Title Progression-Free Survival (PFS) as Assessed by Investigator Using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (v1.1) at Primary Analysis
    Description PFS was defined as the time from randomization to disease progression as determined by the investigator with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 millimeters (mm). For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Median (95% Confidence Interval) [months]
    5.39
    7.43
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0037
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.70
    Confidence Interval (2-Sided) 95%
    0.56 to 0.89
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    2. Primary Outcome
    Title PFS as Assessed by Investigator Using RECIST v1.1 at Final Analysis
    Description PFS was defined as the time from randomization to disease progression as determined by the investigator with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Median (95% Confidence Interval) [months]
    5.55
    8.05
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0008
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.71
    Confidence Interval (2-Sided) 95%
    0.58 to 0.87
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    3. Secondary Outcome
    Title Percentage of Participants With Objective Response (Partial Response [PR] Plus Complete Response [CR]), as Assessed Using RECIST v.1.1 at Primary Analysis
    Description PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions).
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors and measurable disease at baseline, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 134 264
    Number (95% Confidence Interval) [percentage of participants]
    11.9
    5.6%
    28.0
    6.7%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0002
    Comments
    Method Cochran-Mantel-Haenszel
    Comments
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 3.0
    Confidence Interval (2-Sided) 95%
    1.6 to 5.4
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    4. Secondary Outcome
    Title Percentage of Participants With Objective Response (PR Plus CR), as Assessed Using RECIST v.1.1 at Final Analysis
    Description PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions).
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors and measurable disease at baseline, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 134 264
    Number (95% Confidence Interval) [percentage of participants]
    13.4
    6.3%
    31.1
    7.5%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value <0.0001
    Comments
    Method Cochran-Mantel-Haenszel
    Comments
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 3.1
    Confidence Interval (2-Sided) 95%
    1.7 to 5.4
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    5. Secondary Outcome
    Title Overall Survival (OS) at Primary Analysis
    Description OS was defined as the time from the date of randomization to the date of death due to any cause.
    Time Frame From randomization up to death from any cause (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Median (95% Confidence Interval) [months]
    23.56
    26.81
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.4151
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.85
    Confidence Interval (2-Sided) 95%
    0.58 to 1.25
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    6. Secondary Outcome
    Title OS at Final Analysis
    Description OS was defined as the time from the date of randomization to the date of death due to any cause.
    Time Frame From randomization up to death from any cause (up to approximately 6.2 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Median (95% Confidence Interval) [months]
    27.93
    27.79
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.9974
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.00
    Confidence Interval (2-Sided) 95%
    0.75 to 1.33
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    7. Secondary Outcome
    Title Percentage of Participants With Clinical Benefit, as Assessed According to RECIST v1.1 at Primary Analysis
    Description Clinical benefit was defined as objective response (PR+CR), or no disease progression lasting for more than or equal to (>/=) 24 weeks since randomization. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors and measurable disease at baseline, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 134 264
    Number (95% Confidence Interval) [percentage of participants]
    37.3
    17.4%
    51.5
    12.4%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 1.9
    Confidence Interval (2-Sided) 95%
    1.2 to 2.9
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    8. Secondary Outcome
    Title Percentage of Participants With Clinical Benefit, as Assessed According to RECIST v1.1 at Final Analysis
    Description Clinical benefit was defined as objective response (PR+CR), or no disease progression lasting for more than or equal to (>/=) 24 weeks since randomization. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors and measurable disease at baseline, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 134 264
    Number (95% Confidence Interval) [percentage of participants]
    45.5
    21.3%
    59.5
    14.3%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Odds Ratio (OR)
    Estimated Value 1.9
    Confidence Interval (2-Sided) 95%
    1.2 to 3.0
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    9. Secondary Outcome
    Title Duration of Objective Response, as Assessed by Investigator Using RECIST v1.1 at Primary Analysis
    Description Duration of objective response: the time from the first tumor assessment that supported the participant's objective response (CR or PR, whichever was first recorded) to first documented disease progression or death due to any cause, whichever occurred first. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. Disease progression: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame Time from the first occurrence of a documented objective response to the time of the first documented disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors and measurable disease at baseline, regardless of whether they received any amount of study treatment. Data are reported for participants with responses.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 16 74
    Median (95% Confidence Interval) [months]
    7.23
    8.74
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.6708
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.77
    Confidence Interval (2-Sided) 95%
    0.23 to 2.59
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    10. Secondary Outcome
    Title Duration of Objective Response, as Assessed by Investigator Using RECIST v1.1 at Final Analysis
    Description Duration of objective response: the time from the first tumor assessment that supported the participant's objective response (CR or PR, whichever was first recorded) to first documented disease progression or death due to any cause, whichever occurred first. CR was defined as disappearance of all target and non-target lesions and normalization of tumor marker levels (as applicable to non-target lesions). PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. Disease progression: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame Time from the first occurrence of a documented objective response to the time of the first documented disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors and measurable disease at baseline, regardless of whether they received any amount of study treatment. Data are reported for participants with responses.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 18 82
    Median (95% Confidence Interval) [months]
    7.39
    8.97
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.8286
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 1.09
    Confidence Interval (2-Sided) 95%
    0.51 to 2.35
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    11. Secondary Outcome
    Title PFS as Assessed by Blinded Independent Central Review (BICR) Using RECIST v1.1 at Primary Analysis
    Description PFS was defined as the time from randomization to disease progression as determined by BICR with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to the 15 Oct 2017 data cutoff, approximately 2.5 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Median (95% Confidence Interval) [months]
    5.39
    8.97
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0023
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.66
    Confidence Interval (2-Sided) 95%
    0.51 to 0.86
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    12. Secondary Outcome
    Title PFS as Assessed by BICR Using RECIST v1.1 at Final Analysis
    Description PFS was defined as the time from randomization to disease progression as determined by BICR with the use of RECIST v1.1 or death due to any cause, whichever occurred earlier. Disease progression was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, including baseline. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. For non-target lesions, disease progression was defined as unequivocal progression of existing lesions. The appearance of one or more new lesions was also considered progression.
    Time Frame From randomization until the first occurrence of disease progression or death from any cause, whichever occurs earlier (up to approximately 6.2 years)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Median (95% Confidence Interval) [months]
    5.55
    9.20
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Placebo+Fulvestrant, Taselisib+Fulvestrant
    Comments
    Type of Statistical Test Superiority
    Comments
    Statistical Test of Hypothesis p-Value 0.0095
    Comments
    Method Log Rank
    Comments
    Method of Estimation Estimation Parameter Hazard Ratio (HR)
    Estimated Value 0.72
    Confidence Interval (2-Sided) 95%
    0.56 to 0.92
    Parameter Dispersion Type:
    Value:
    Estimation Comments
    13. Secondary Outcome
    Title Percentage of Participants With Adverse Events at Primary Analysis
    Description An adverse event was any untoward medical occurrence in a clinical investigation participant administered a pharmaceutical product, regardless of causal attribution.
    Time Frame From randomization up to the 15 Oct 2017 data cutoff, approximately 2.5 years.

    Outcome Measure Data

    Analysis Population Description
    The safety-evaluable population included all randomized participants who received at least one dose of taselisib or placebo or fulvestrant.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 213 416
    Number [percentage of participants]
    89.7
    41.9%
    95.4
    22.9%
    14. Secondary Outcome
    Title Percentage of Participants With Adverse Events at Final Analysis
    Description An adverse event was any untoward medical occurrence in a clinical investigation participant administered a pharmaceutical product, regardless of causal attribution.
    Time Frame From randomization up to approximately 6.2 years

    Outcome Measure Data

    Analysis Population Description
    The safety-evaluable population included all randomized participants who received at least one dose of taselisib or placebo or fulvestrant.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 213 416
    Number [percentage of participants]
    91.1
    42.6%
    97.1
    23.3%
    15. Secondary Outcome
    Title Maximum Observed Plasma Concentration (Cmax) of Taselisib
    Description
    Time Frame 1 to 4 hours (hrs) post-dose on Cycle (C) 1, Day (D) 1; 0 to 3 hrs pre-dose and 2 to 6 hrs post dose on Cycle 2, Day 1 (each cycle=28 days)

    Outcome Measure Data

    Analysis Population Description
    The Pharmacokinetic (PK) population included all participants who received at least one dose of taselisib and provided valid (adequately documented dose time and PK sample time) PK assessments.
    Arm/Group Title Taselisib+Fulvestrant
    Arm/Group Description Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 417
    C1D1
    18.2
    (14.6)
    C2D1
    66.6
    (35.2)
    16. Secondary Outcome
    Title Minimum Observed Plasma Concentration (Cmin) of Taselisib
    Description
    Time Frame 1 to 4 hrs post-dose on Cycle 1, Day 1; 0 to 3 hrs pre-dose and 2 to 6 hrs post dose on Cycle 2, Day 1; 0 to 3 hrs pre-dose on Cycle 6, Day 1 (each cycle=28 days)

    Outcome Measure Data

    Analysis Population Description
    The PK population included all participants who received at least one dose of taselisib and provided valid (adequately documented dose time and PK sample time) PK assessments.
    Arm/Group Title Taselisib+Fulvestrant
    Arm/Group Description Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 417
    C2D1
    42.8
    (26.6)
    C6D1
    35.3
    (31.5)
    17. Secondary Outcome
    Title Change From Baseline in European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Core 30 (QLQ-C30) Score
    Description The EORTC QLQ-C30 consists of 30 questions that comprise aspects of participant's functioning assessment (physical, emotional, role, cognitive, and social); symptom scales (fatigue; nausea, vomiting, and pain; the global health/quality of life [QoL]); and single items (dyspnoea, insomnia, appetite loss, constipation, diarrhoea, and financial difficulties), within a recall period of "the past week." Most questions used a 4-point scale (1=Not at all to 4=Very much; two questions used a 7-point scale (1=Very poor to 7=Excellent). Scores were averaged and transformed to a 0-100 scale; a higher score for Global Qol/functional scales=better level of functioning; a higher score for symptom scale=greater degree of symptoms.
    Time Frame Baseline, C2D1 up to C7D1 (each cycle=28 days)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment. Data are reported for evaluable participants.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Appetite Loss: Baseline
    15.9
    (25.9)
    15.3
    (23.3)
    Appetite Loss: Change at C2D1
    -0.2
    (25.7)
    6.2
    (26.2)
    Appetite Loss: Change at C3D1
    -4.3
    (27.2)
    8.7
    (28.1)
    Appetite Loss: Change at C4D1
    -1.7
    (26.1)
    8.4
    (28.1)
    Appetite Loss: Change at C5D1
    -0.4
    (26.6)
    6.0
    (27.2)
    Appetite Loss: Change at C6D1
    -0.5
    (22.3)
    6.6
    (27.2)
    Appetite Loss: Change at C7D1
    -5.7
    (28.7)
    4.6
    (26.5)
    Cognitive Functioning: Baseline
    85.3
    (18.1)
    85.9
    (18.7)
    Cognitive Functioning: Change at C2D1
    0.7
    (16.5)
    -1.1
    (16.8)
    Cognitive Functioning: Change at C3D1
    0.9
    (17.9)
    -2.9
    (19.1)
    Cognitive Functioning: Change at C4D1
    1.8
    (18.9)
    -1.7
    (18.4)
    Cognitive Functioning: Change at C5D1
    -1.5
    (17.7)
    -3.2
    (17.7)
    Cognitive Functioning: Change at C6D1
    -0.5
    (18.5)
    -2.8
    (17.6)
    Cognitive Functioning: Change at C7D1
    -2.3
    (18.1)
    -0.8
    (18.8)
    Constipation: Baseline
    15.8
    (23.9)
    14.5
    (23.7)
    Constipation: Change at C2D1
    0.0
    (24.0)
    -6.0
    (21.1)
    Constipation: Change at C3D1
    -2.3
    (25.0)
    -4.9
    (20.8)
    Constipation: Change at C4D1
    -2.7
    (24.6)
    -5.5
    (22.0)
    Constipation: Change at C5D1
    -1.3
    (22.7)
    -4.0
    (21.0)
    Constipation: Change at C6D1
    -3.0
    (27.3)
    -3.5
    (21.8)
    Constipation: Change at C7D1
    -4.6
    (24.5)
    -6.9
    (22.5)
    Diarrhoea: Baseline
    6.3
    (15.1)
    5.0
    (14.4)
    Diarrhoea: Change at C2D1
    -0.5
    (17.9)
    10.2
    (22.4)
    Diarrhoea: Change at C3D1
    -0.9
    (16.6)
    13.7
    (26.3)
    Diarrhoea: Change at C4D1
    -2.7
    (21.0)
    13.1
    (28.9)
    Diarrhoea: Change at C5D1
    -2.6
    (19.4)
    11.1
    (27.7)
    Diarrhoea: Change at C6D1
    -2.0
    (17.4)
    14.0
    (29.1)
    Diarrhoea: Change at C7D1
    1.1
    (20.7)
    15.6
    (30.9)
    Dyspnoea: Baseline
    15.0
    (23.0)
    15.4
    (22.2)
    Dyspnoea: Change at C2D1
    4.1
    (23.5)
    -2.1
    (19.5)
    Dyspnoea: Change at C3D1
    2.8
    (20.3)
    0.0
    (20.9)
    Dyspnoea: Change at C4D1
    0.7
    (23.2)
    0.0
    (23.3)
    Dyspnoea: Change at C5D1
    2.6
    (26.5)
    1.6
    (23.2)
    Dyspnoea: Change at C6D1
    3.0
    (26.6)
    0.2
    (23.4)
    Dyspnoea: Change at C7D1
    1.7
    (24.5)
    -2.1
    (21.2)
    Emotional Functioning: Baseline
    73.1
    (22.1)
    71.9
    (22.1)
    Emotional Functioning: Change at C2D1
    4.0
    (17.9)
    5.1
    (18.5)
    Emotional Functioning: Change at C3D1
    4.5
    (19.1)
    2.3
    (21.6)
    Emotional Functioning: Change at C4D1
    4.5
    (18.5)
    2.6
    (19.7)
    Emotional Functioning: Change at C5D1
    5.2
    (20.1)
    -0.4
    (21.8)
    Emotional Functioning: Change at C6D1
    2.1
    (20.5)
    2.4
    (21.6)
    Emotional Functioning: Change at C7D1
    5.3
    (20.7)
    2.8
    (19.7)
    Fatigue: Baseline
    30.8
    (22.5)
    30.8
    (22.0)
    Fatigue: Change at C2D1
    2.0
    (19.2)
    -0.5
    (18.8)
    Fatigue: Change at C3D1
    -1.5
    (19.5)
    1.8
    (21.5)
    Fatigue: Change at C4D1
    -0.2
    (19.1)
    2.4
    (21.9)
    Fatigue: Change at C5D1
    -0.1
    (20.8)
    2.8
    (20.4)
    Fatigue: Change at C6D1
    3.3
    (20.0)
    2.4
    (20.5)
    Fatigue: Change at C7D1
    1.0
    (20.0)
    1.8
    (20.5)
    Financial Difficulties: Baseline
    18.5
    (25.8)
    19.2
    (27.1)
    Financial Difficulties: Change at C2D1
    -1.1
    (20.4)
    -2.8
    (21.4)
    Financial Difficulties: Change at C3D1
    -0.9
    (24.9)
    -1.6
    (24.4)
    Financial Difficulties: Change at C4D1
    0.7
    (25.1)
    -0.3
    (23.7)
    Financial Difficulties: Change at C5D1
    -0.4
    (28.0)
    0.3
    (24.9)
    Financial Difficulties: Change at C6D1
    5.6
    (30.1)
    0.4
    (23.6)
    Financial Difficulties: Change at C7D1
    -0.6
    (26.1)
    2.1
    (23.1)
    Global Health Status/ QoL: Baseline
    65.2
    (18.4)
    67.4
    (20.3)
    Global Health Status/ QoL: Change at C2D1
    -0.1
    (16.7)
    1.0
    (19.9)
    Global Health Status/ QoL: Change at C3D1
    -1.0
    (18.5)
    -1.5
    (20.9)
    Global Health Status/ QoL: Change at C4D1
    -1.5
    (18.6)
    -1.6
    (20.3)
    Global Health Status/ QoL: Change at C5D1
    0.3
    (19.9)
    -2.8
    (20.3)
    Global Health Status/ QoL: Change at C6D1
    -1.6
    (18.6)
    -3.4
    (19.5)
    Global Health Status/ QoL: Change at C7D1
    -1.1
    (18.9)
    -1.0
    (18.9)
    Insomnia: Baseline
    26.0
    (27.6)
    26.5
    (27.7)
    Insomnia: Change at C2D1
    -0.9
    (24.4)
    -3.8
    (23.7)
    Insomnia: Change at C3D1
    -3.4
    (30.1)
    -4.1
    (26.4)
    Insomnia: Change at C4D1
    -4.0
    (28.9)
    -1.0
    (26.8)
    Insomnia: Change at C5D1
    -0.4
    (29.1)
    -3.9
    (25.6)
    Insomnia: Change at C6D1
    -2.0
    (30.3)
    -2.4
    (28.2)
    Insomnia: Change at C7D1
    -4.1
    (28.2)
    -1.8
    (27.2)
    Nausea/Vomiting: Baseline
    5.9
    (13.4)
    6.7
    (13.7)
    Nausea/Vomiting: Change at C2D1
    0.1
    (11.9)
    1.6
    (18.1)
    Nausea/Vomiting: Change at C3D1
    0.7
    (15.2)
    2.2
    (17.6)
    Nausea/Vomiting: Change at C4D1
    0.3
    (17.9)
    2.3
    (18.6)
    Nausea/Vomiting: Change at C5D1
    -1.1
    (18.3)
    0.5
    (15.4)
    Nausea/Vomiting: Change at C6D1
    1.5
    (18.0)
    -0.6
    (15.9)
    Nausea/Vomiting: Change at C7D1
    2.6
    (15.5)
    2.2
    (18.8)
    Pain: Baseline
    28.0
    (25.4)
    27.1
    (24.9)
    Pain: Change at C2D1
    -0.2
    (24.0)
    -5.0
    (20.8)
    Pain: Change at C3D1
    -3.7
    (23.3)
    -3.5
    (22.6)
    Pain: Change at C4D1
    -3.2
    (24.7)
    -1.7
    (24.2)
    Pain: Change at C5D1
    -3.3
    (24.2)
    -4.3
    (22.9)
    Pain: Change at C6D1
    -1.0
    (23.0)
    -2.2
    (22.5)
    Pain: Change at C7D1
    0.3
    (23.5)
    -4.4
    (19.8)
    Physical Functioning: Baseline
    76.7
    (19.9)
    78.4
    (18.8)
    Physical Functioning: Change at C2D1
    -1.1
    (13.4)
    1.6
    (12.7)
    Physical Functioning: Change at C3D1
    2.0
    (14.1)
    0.8
    (15.7)
    Physical Functioning: Change at C4D1
    1.5
    (16.1)
    0.3
    (15.7)
    Physical Functioning: Change at C5D1
    2.0
    (17.7)
    1.0
    (13.5)
    Physical Functioning: Change at C6D1
    0.9
    (18.4)
    1.1
    (14.9)
    Physical Functioning: Change at C7D1
    1.6
    (16.2)
    0.6
    (14.2)
    Role Functioning: Baseline
    79.1
    (24.6)
    78.7
    (24.0)
    Role Functioning: Change at C2D1
    -2.0
    (19.3)
    1.7
    (21.5)
    Role Functioning: Change at C3D1
    -0.4
    (23.4)
    -1.0
    (23.9)
    Role Functioning: Change at C4D1
    0.3
    (23.0)
    0.4
    (24.2)
    Role Functioning: Change at C5D1
    1.8
    (22.5)
    -1.6
    (21.6)
    Role Functioning: Change at C6D1
    -1.3
    (23.8)
    -1.6
    (23.4)
    Role Functioning: Change at C7D1
    -0.3
    (22.6)
    0.0
    (22.9)
    Social Functioning: Baseline
    83.2
    (21.8)
    81.2
    (23.1)
    Social Functioning: Change at C2D1
    -0.8
    (19.9)
    2.7
    (20.0)
    Social Functioning: Change at C3D1
    1.3
    (21.9)
    -0.8
    (23.2)
    Social Functioning: Change at C4D1
    1.8
    (18.2)
    -0.5
    (21.3)
    Social Functioning: Change at C5D1
    0.9
    (19.4)
    -1.0
    (23.3)
    Social Functioning: Change at C6D1
    -0.8
    (20.1)
    -1.6
    (23.4)
    Social Functioning: Change at C7D1
    0.6
    (21.2)
    0.1
    (19.7)
    18. Secondary Outcome
    Title Change From Baseline in Modified EORTC Quality of Life Questionnaire Breast Cancer Module 23 (QLQ-BR23) Score
    Description EORTC-QLQ-BR23 is a 23-item breast cancer-specific companion module to the EORTC-QLQ-C30 and consists of functional scales (body image, sexual enjoyment, sexual functioning, future perspective [FP]) and symptom scales (systemic side effects [SE], upset by hair loss, arm symptoms, breast symptoms). Questions used a 4-point scale (1=not at all, 2=a little, 3=quite a bit, 4=very much). Scores were averaged and transformed to a 0-100 scale. Higher scores for the functional scales indicated a higher/better level of functioning/healthy functioning. Higher scores for the symptom scales indicated worse symptoms.
    Time Frame Baseline, C2D1 up to C7D1 (each cycle=28 days)

    Outcome Measure Data

    Analysis Population Description
    Randomized participants with PIK3CA-mutant tumors, regardless of whether they received any amount of study treatment. Data are reported for evaluable participants.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    Measure Participants 176 340
    Arm Symptoms: Baseline
    15.5
    (18.8)
    19.3
    (21.1)
    Arm Symptoms: Change at C2D1
    0.1
    (17.4)
    -5.4
    (15.6)
    Arm Symptoms: Change at C3D1
    -0.7
    (19.5)
    -6.1
    (17.4)
    Arm Symptoms: Change at C4D1
    -1.4
    (21.9)
    -5.8
    (15.8)
    Arm Symptoms: Change at C5D1
    0.8
    (17.0)
    -6.6
    (15.7)
    Arm Symptoms: Change at C6D1
    1.1
    (20.8)
    -5.0
    (18.5)
    Arm Symptoms: Change at C7D1
    -1.2
    (19.7)
    -6.0
    (15.2)
    Body Image: Baseline
    82.0
    (21.7)
    80.8
    (22.5)
    Body Image: Change at C2D1
    1.8
    (14.9)
    1.5
    (16.5)
    Body Image: Change at C3D1
    1.6
    (18.0)
    1.1
    (20.6)
    Body Image: Change at C4D1
    1.1
    (17.8)
    1.8
    (20.6)
    Body Image: Change at C5D1
    0.2
    (19.5)
    0.0
    (17.5)
    Body Image: Change at C6D1
    1.1
    (21.0)
    0.4
    (18.2)
    Body Image: Change at C7D1
    5.6
    (18.8)
    -0.3
    (18.2)
    Breast Symptoms: Baseline
    8.7
    (14.6)
    11.0
    (14.1)
    Breast Symptoms: Change at C2D1
    0.0
    (13.7)
    -3.0
    (11.8)
    Breast Symptoms: Change at C3D1
    -0.8
    (13.3)
    -3.5
    (11.6)
    Breast Symptoms: Change at C4D1
    0.1
    (14.2)
    -3.0
    (11.8)
    Breast Symptoms: Change at C5D1
    -0.9
    (14.5)
    -2.0
    (11.7)
    Breast Symptoms: Change at C6D1
    1.7
    (16.0)
    -2.0
    (12.9)
    Breast Symptoms: Change at C7D1
    0.3
    (12.6)
    -3.2
    (13.1)
    Future Perspective: Baseline
    47.4
    (31.5)
    47.3
    (29.6)
    Future Perspective: Change at C2D1
    3.4
    (30.1)
    6.8
    (27.4)
    Future Perspective: Change at C3D1
    5.1
    (29.5)
    4.3
    (30.5)
    Future Perspective: Change at C4D1
    6.0
    (30.7)
    7.0
    (29.2)
    Future Perspective: Change at C5D1
    6.0
    (30.3)
    5.0
    (26.8)
    Future Perspective: Change at C6D1
    6.9
    (28.8)
    9.0
    (31.4)
    Future Perspective: Change at C7D1
    12.9
    (29.4)
    7.0
    (30.2)
    Sexual Enjoyment: Baseline
    51.9
    (26.7)
    62.8
    (26.6)
    Sexual Enjoyment: Change at C2D1
    5.9
    (21.2)
    0.0
    (18.1)
    Sexual Enjoyment: Change at C3D1
    4.8
    (22.1)
    1.3
    (24.0)
    Sexual Enjoyment: Change at C4D1
    -8.3
    (15.4)
    4.2
    (24.7)
    Sexual Enjoyment: Change at C5D1
    -4.8
    (23.0)
    2.6
    (29.7)
    Sexual Enjoyment: Change at C6D1
    -6.7
    (14.9)
    -5.9
    (24.3)
    Sexual Enjoyment: Change at C7D1
    -13.3
    (29.8)
    9.5
    (20.4)
    Sexual Functioning: Baseline
    89.6
    (17.9)
    89.9
    (17.3)
    Sexual Functioning: Change at C2D1
    1.6
    (11.0)
    1.5
    (14.2)
    Sexual Functioning: Change at C3D1
    1.4
    (10.6)
    1.8
    (15.6)
    Sexual Functioning: Change at C4D1
    1.9
    (15.4)
    1.6
    (13.9)
    Sexual Functioning: Change at C5D1
    -0.8
    (20.6)
    2.6
    (15.1)
    Sexual Functioning: Change at C6D1
    2.4
    (18.8)
    2.7
    (15.0)
    Sexual Functioning: Change at C7D1
    2.7
    (18.1)
    2.3
    (15.7)
    Systematic Therapy SEs: Baseline
    15.7
    (14.2)
    14.7
    (12.0)
    Systematic Therapy SEs: Change at C2D1
    0.2
    (11.4)
    2.5
    (11.0)
    Systematic Therapy SEs: Change at C3D1
    1.2
    (14.4)
    4.0
    (13.6)
    Systematic Therapy SEs: Change at C4D1
    1.5
    (13.2)
    4.0
    (13.3)
    Systematic Therapy SEs: Change at C5D1
    2.4
    (13.8)
    5.5
    (14.1)
    Systematic Therapy SEs: Change at C6D1
    2.9
    (15.2)
    5.7
    (15.6)
    Systematic Therapy SEs: Change at C7D1
    3.5
    (16.7)
    4.8
    (15.0)
    Upset by Hair Loss: Baseline
    23.2
    (28.2)
    27.0
    (29.2)
    Upset by Hair Loss: Change at C2D1
    -11.9
    (28.1)
    -4.3
    (22.3)
    Upset by Hair Loss: Change at C3D1
    -7.7
    (30.9)
    0.0
    (26.7)
    Upset by Hair Loss: Change at C4D1
    2.6
    (16.5)
    0.0
    (29.2)
    Upset by Hair Loss: Change at C5D1
    0.0
    (25.2)
    10.7
    (27.3)
    Upset by Hair Loss: Change at C6D1
    11.1
    (27.2)
    16.7
    (30.2)
    Upset by Hair Loss: Change at C7D1
    4.2
    (11.8)
    14.1
    (32.9)

    Adverse Events

    Time Frame From randomization up to the end of study, approximately 6.2 years
    Adverse Event Reporting Description All-cause mortality was reported based on the ITT population: all randomized participants regardless of whether they received any amount of study treatment. Serious and non-serious adverse events were reported based on the safety-evaluable population: all randomized participants who received at least one dose of taselisib, placebo or fulvestrant regardless of the PIK3CA-mutation status of their tumors with participants allocated to the treatment arm associated with the regimen actually received.
    Arm/Group Title Placebo+Fulvestrant Taselisib+Fulvestrant
    Arm/Group Description Participants received placebo taken orally QD beginning at Cycle 1, Day 1, and fulvestrant 500 mg administered by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor. Participants received taselisib 4 mg taken orally QD beginning at Cycle 1, Day 1 and fulvestrant 500 mg by IM injection at Cycle 1, Days 1 and 15, and then on Day 1 of each subsequent 28-day cycle until disease progression, unacceptable toxicity, or study termination by the Sponsor.
    All Cause Mortality
    Placebo+Fulvestrant Taselisib+Fulvestrant
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 100/214 (46.7%) 197/417 (47.2%)
    Serious Adverse Events
    Placebo+Fulvestrant Taselisib+Fulvestrant
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 23/213 (10.8%) 154/416 (37%)
    Blood and lymphatic system disorders
    ANAEMIA 1/213 (0.5%) 2 2/416 (0.5%) 2
    PANCYTOPENIA 0/213 (0%) 0 1/416 (0.2%) 1
    Cardiac disorders
    ATRIAL FIBRILLATION 0/213 (0%) 0 2/416 (0.5%) 2
    CARDIAC FAILURE CONGESTIVE 0/213 (0%) 0 1/416 (0.2%) 1
    MYOCARDIAL INFARCTION 0/213 (0%) 0 1/416 (0.2%) 1
    SUPRAVENTRICULAR TACHYCARDIA 0/213 (0%) 0 1/416 (0.2%) 1
    Gastrointestinal disorders
    ABDOMINAL PAIN 0/213 (0%) 0 1/416 (0.2%) 1
    ABDOMINAL PAIN UPPER 0/213 (0%) 0 1/416 (0.2%) 1
    ALCOHOLIC PANCREATITIS 0/213 (0%) 0 1/416 (0.2%) 1
    COLITIS 0/213 (0%) 0 16/416 (3.8%) 17
    DIARRHOEA 0/213 (0%) 0 37/416 (8.9%) 40
    DYSPEPSIA 0/213 (0%) 0 1/416 (0.2%) 1
    DYSPHAGIA 0/213 (0%) 0 1/416 (0.2%) 1
    ENTERITIS 0/213 (0%) 0 1/416 (0.2%) 1
    ENTEROCOLITIS 0/213 (0%) 0 3/416 (0.7%) 3
    GASTRIC ULCER 0/213 (0%) 0 1/416 (0.2%) 1
    GASTRITIS 0/213 (0%) 0 2/416 (0.5%) 2
    NAUSEA 2/213 (0.9%) 2 4/416 (1%) 5
    OESOPHAGEAL ULCER 0/213 (0%) 0 1/416 (0.2%) 1
    OESOPHAGITIS 0/213 (0%) 0 1/416 (0.2%) 1
    PANCREATITIS 0/213 (0%) 0 1/416 (0.2%) 1
    PANCREATITIS ACUTE 0/213 (0%) 0 2/416 (0.5%) 2
    SMALL INTESTINAL OBSTRUCTION 0/213 (0%) 0 2/416 (0.5%) 3
    STOMATITIS 0/213 (0%) 0 2/416 (0.5%) 2
    VOMITING 1/213 (0.5%) 1 4/416 (1%) 4
    General disorders
    ASTHENIA 0/213 (0%) 0 1/416 (0.2%) 1
    CHEST DISCOMFORT 0/213 (0%) 0 2/416 (0.5%) 2
    CHEST PAIN 0/213 (0%) 0 3/416 (0.7%) 3
    CHILLS 1/213 (0.5%) 1 0/416 (0%) 0
    DEATH 1/213 (0.5%) 1 3/416 (0.7%) 3
    FATIGUE 1/213 (0.5%) 1 2/416 (0.5%) 2
    MUCOSAL INFLAMMATION 0/213 (0%) 0 1/416 (0.2%) 1
    PAIN 1/213 (0.5%) 1 0/416 (0%) 0
    PYREXIA 2/213 (0.9%) 2 3/416 (0.7%) 3
    SWELLING FACE 1/213 (0.5%) 1 0/416 (0%) 0
    Hepatobiliary disorders
    CHOLECYSTITIS ACUTE 0/213 (0%) 0 1/416 (0.2%) 1
    CHOLELITHIASIS 0/213 (0%) 0 1/416 (0.2%) 1
    HEPATOTOXICITY 0/213 (0%) 0 1/416 (0.2%) 1
    Immune system disorders
    ANAPHYLACTIC SHOCK 0/213 (0%) 0 1/416 (0.2%) 1
    Infections and infestations
    APPENDICITIS PERFORATED 0/213 (0%) 0 1/416 (0.2%) 1
    ATYPICAL MYCOBACTERIAL PNEUMONIA 0/213 (0%) 0 1/416 (0.2%) 1
    BRONCHITIS 0/213 (0%) 0 1/416 (0.2%) 1
    CELLULITIS 0/213 (0%) 0 2/416 (0.5%) 2
    CHOLERA 0/213 (0%) 0 1/416 (0.2%) 1
    CLOSTRIDIUM DIFFICILE COLITIS 0/213 (0%) 0 1/416 (0.2%) 1
    CYSTITIS ESCHERICHIA 0/213 (0%) 0 1/416 (0.2%) 1
    DIARRHOEA INFECTIOUS 0/213 (0%) 0 3/416 (0.7%) 3
    ENTEROCOLITIS INFECTIOUS 0/213 (0%) 0 2/416 (0.5%) 2
    GASTROENTERITIS 0/213 (0%) 0 1/416 (0.2%) 1
    GASTROENTERITIS VIRAL 0/213 (0%) 0 1/416 (0.2%) 1
    HERPES ZOSTER 0/213 (0%) 0 1/416 (0.2%) 1
    LOWER RESPIRATORY TRACT INFECTION 0/213 (0%) 0 1/416 (0.2%) 1
    PNEUMONIA 1/213 (0.5%) 1 10/416 (2.4%) 10
    RESPIRATORY TRACT INFECTION 0/213 (0%) 0 1/416 (0.2%) 1
    SEPSIS 1/213 (0.5%) 1 5/416 (1.2%) 5
    SEPTIC ARTHRITIS STAPHYLOCOCCAL 0/213 (0%) 0 1/416 (0.2%) 2
    SKIN INFECTION 0/213 (0%) 0 2/416 (0.5%) 2
    URINARY TRACT INFECTION 0/213 (0%) 0 3/416 (0.7%) 3
    UROSEPSIS 0/213 (0%) 0 1/416 (0.2%) 1
    VIRAL INFECTION 0/213 (0%) 0 1/416 (0.2%) 1
    Injury, poisoning and procedural complications
    CLAVICLE FRACTURE 0/213 (0%) 0 1/416 (0.2%) 1
    FALL 1/213 (0.5%) 1 2/416 (0.5%) 2
    FEMORAL NECK FRACTURE 1/213 (0.5%) 1 0/416 (0%) 0
    FEMUR FRACTURE 0/213 (0%) 0 1/416 (0.2%) 1
    HIP FRACTURE 1/213 (0.5%) 1 0/416 (0%) 0
    JOINT DISLOCATION 0/213 (0%) 0 1/416 (0.2%) 1
    TOXICITY TO VARIOUS AGENTS 0/213 (0%) 0 1/416 (0.2%) 1
    WRIST FRACTURE 1/213 (0.5%) 1 0/416 (0%) 0
    Investigations
    ALANINE AMINOTRANSFERASE INCREASED 1/213 (0.5%) 1 2/416 (0.5%) 2
    ASPARTATE AMINOTRANSFERASE INCREASED 1/213 (0.5%) 1 1/416 (0.2%) 1
    WHITE BLOOD CELL COUNT DECREASED 1/213 (0.5%) 1 0/416 (0%) 0
    Metabolism and nutrition disorders
    DECREASED APPETITE 0/213 (0%) 0 1/416 (0.2%) 1
    DEHYDRATION 1/213 (0.5%) 1 7/416 (1.7%) 7
    HYPERGLYCAEMIA 0/213 (0%) 0 7/416 (1.7%) 7
    Musculoskeletal and connective tissue disorders
    ARTHRALGIA 0/213 (0%) 0 2/416 (0.5%) 3
    BACK PAIN 0/213 (0%) 0 1/416 (0.2%) 1
    COCCYDYNIA 0/213 (0%) 0 1/416 (0.2%) 1
    MUSCLE SPASMS 1/213 (0.5%) 2 0/416 (0%) 0
    MUSCULAR WEAKNESS 1/213 (0.5%) 1 0/416 (0%) 0
    OSTEONECROSIS OF JAW 1/213 (0.5%) 1 0/416 (0%) 0
    RHABDOMYOLYSIS 0/213 (0%) 0 1/416 (0.2%) 1
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    HEPATOCELLULAR CARCINOMA 0/213 (0%) 0 1/416 (0.2%) 1
    INTRACRANIAL TUMOUR HAEMORRHAGE 0/213 (0%) 0 1/416 (0.2%) 1
    Nervous system disorders
    CEREBRAL HAEMATOMA 0/213 (0%) 0 1/416 (0.2%) 1
    CEREBROVASCULAR ACCIDENT 2/213 (0.9%) 2 1/416 (0.2%) 1
    FACIAL PARALYSIS 1/213 (0.5%) 1 0/416 (0%) 0
    HEADACHE 0/213 (0%) 0 3/416 (0.7%) 3
    HYPOAESTHESIA 0/213 (0%) 0 2/416 (0.5%) 2
    TRANSIENT ISCHAEMIC ATTACK 0/213 (0%) 0 1/416 (0.2%) 1
    TRIGEMINAL NEURALGIA 0/213 (0%) 0 1/416 (0.2%) 1
    VITH NERVE PARALYSIS 1/213 (0.5%) 1 0/416 (0%) 0
    Psychiatric disorders
    CONFUSIONAL STATE 0/213 (0%) 0 1/416 (0.2%) 1
    MENTAL STATUS CHANGES 0/213 (0%) 0 2/416 (0.5%) 2
    Renal and urinary disorders
    ACUTE KIDNEY INJURY 0/213 (0%) 0 4/416 (1%) 4
    HAEMATURIA 0/213 (0%) 0 1/416 (0.2%) 1
    RENAL FAILURE 0/213 (0%) 0 1/416 (0.2%) 1
    URETEROLITHIASIS 1/213 (0.5%) 1 0/416 (0%) 0
    Reproductive system and breast disorders
    INTERMENSTRUAL BLEEDING 0/213 (0%) 0 1/416 (0.2%) 1
    Respiratory, thoracic and mediastinal disorders
    ACUTE RESPIRATORY DISTRESS SYNDROME 0/213 (0%) 0 1/416 (0.2%) 1
    ACUTE RESPIRATORY FAILURE 0/213 (0%) 0 1/416 (0.2%) 1
    COUGH 0/213 (0%) 0 1/416 (0.2%) 1
    DYSPNOEA 1/213 (0.5%) 1 1/416 (0.2%) 1
    INTERSTITIAL LUNG DISEASE 0/213 (0%) 0 1/416 (0.2%) 1
    PLEURAL EFFUSION 2/213 (0.9%) 2 1/416 (0.2%) 2
    PLEURITIC PAIN 1/213 (0.5%) 1 0/416 (0%) 0
    PNEUMONITIS 1/213 (0.5%) 1 10/416 (2.4%) 10
    PNEUMOTHORAX 0/213 (0%) 0 2/416 (0.5%) 2
    PULMONARY EMBOLISM 0/213 (0%) 0 1/416 (0.2%) 1
    PULMONARY OEDEMA 1/213 (0.5%) 1 0/416 (0%) 0
    RESPIRATORY FAILURE 0/213 (0%) 0 1/416 (0.2%) 1
    Skin and subcutaneous tissue disorders
    DRUG ERUPTION 0/213 (0%) 0 1/416 (0.2%) 1
    PRURITUS 0/213 (0%) 0 1/416 (0.2%) 1
    RASH 0/213 (0%) 0 2/416 (0.5%) 2
    RASH MACULO-PAPULAR 0/213 (0%) 0 1/416 (0.2%) 1
    TOXIC SKIN ERUPTION 0/213 (0%) 0 1/416 (0.2%) 1
    Vascular disorders
    EMBOLISM 0/213 (0%) 0 1/416 (0.2%) 1
    EMBOLISM VENOUS 0/213 (0%) 0 1/416 (0.2%) 1
    HYPERTENSION 0/213 (0%) 0 1/416 (0.2%) 1
    SHOCK 0/213 (0%) 0 1/416 (0.2%) 1
    Other (Not Including Serious) Adverse Events
    Placebo+Fulvestrant Taselisib+Fulvestrant
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 183/213 (85.9%) 396/416 (95.2%)
    Blood and lymphatic system disorders
    ANAEMIA 20/213 (9.4%) 23 44/416 (10.6%) 62
    NEUTROPENIA 9/213 (4.2%) 12 28/416 (6.7%) 39
    Gastrointestinal disorders
    ABDOMINAL PAIN 20/213 (9.4%) 25 55/416 (13.2%) 67
    ABDOMINAL PAIN UPPER 8/213 (3.8%) 8 31/416 (7.5%) 35
    CONSTIPATION 32/213 (15%) 36 32/416 (7.7%) 36
    DIARRHOEA 45/213 (21.1%) 76 255/416 (61.3%) 602
    DRY MOUTH 18/213 (8.5%) 19 55/416 (13.2%) 62
    DYSPEPSIA 5/213 (2.3%) 5 32/416 (7.7%) 51
    GASTROOESOPHAGEAL REFLUX DISEASE 4/213 (1.9%) 4 23/416 (5.5%) 25
    NAUSEA 54/213 (25.4%) 71 154/416 (37%) 233
    STOMATITIS 8/213 (3.8%) 11 87/416 (20.9%) 139
    VOMITING 25/213 (11.7%) 41 86/416 (20.7%) 116
    General disorders
    ASTHENIA 40/213 (18.8%) 65 77/416 (18.5%) 104
    FATIGUE 40/213 (18.8%) 51 105/416 (25.2%) 134
    MUCOSAL INFLAMMATION 11/213 (5.2%) 15 44/416 (10.6%) 67
    OEDEMA PERIPHERAL 11/213 (5.2%) 13 19/416 (4.6%) 25
    PYREXIA 9/213 (4.2%) 10 48/416 (11.5%) 61
    Infections and infestations
    UPPER RESPIRATORY TRACT INFECTION 11/213 (5.2%) 17 33/416 (7.9%) 41
    URINARY TRACT INFECTION 9/213 (4.2%) 10 38/416 (9.1%) 50
    Investigations
    ALANINE AMINOTRANSFERASE INCREASED 9/213 (4.2%) 13 36/416 (8.7%) 47
    ASPARTATE AMINOTRANSFERASE INCREASED 15/213 (7%) 19 35/416 (8.4%) 47
    BLOOD ALKALINE PHOSPHATASE INCREASED 12/213 (5.6%) 12 15/416 (3.6%) 17
    BLOOD CREATININE INCREASED 6/213 (2.8%) 7 25/416 (6%) 28
    WEIGHT DECREASED 6/213 (2.8%) 6 42/416 (10.1%) 50
    Metabolism and nutrition disorders
    DECREASED APPETITE 23/213 (10.8%) 25 119/416 (28.6%) 136
    HYPERGLYCAEMIA 21/213 (9.9%) 26 166/416 (39.9%) 233
    HYPOKALAEMIA 2/213 (0.9%) 2 31/416 (7.5%) 37
    Musculoskeletal and connective tissue disorders
    ARTHRALGIA 35/213 (16.4%) 50 69/416 (16.6%) 95
    BACK PAIN 28/213 (13.1%) 32 62/416 (14.9%) 82
    BONE PAIN 17/213 (8%) 19 24/416 (5.8%) 26
    MUSCLE SPASMS 6/213 (2.8%) 8 32/416 (7.7%) 34
    MYALGIA 14/213 (6.6%) 14 36/416 (8.7%) 42
    PAIN IN EXTREMITY 19/213 (8.9%) 27 33/416 (7.9%) 42
    Nervous system disorders
    DIZZINESS 18/213 (8.5%) 23 46/416 (11.1%) 58
    DYSGEUSIA 5/213 (2.3%) 5 36/416 (8.7%) 41
    HEADACHE 27/213 (12.7%) 38 88/416 (21.2%) 122
    Psychiatric disorders
    INSOMNIA 18/213 (8.5%) 19 37/416 (8.9%) 40
    Respiratory, thoracic and mediastinal disorders
    COUGH 34/213 (16%) 41 64/416 (15.4%) 78
    DYSPNOEA 17/213 (8%) 17 50/416 (12%) 53
    Skin and subcutaneous tissue disorders
    ALOPECIA 6/213 (2.8%) 6 50/416 (12%) 53
    DRY SKIN 10/213 (4.7%) 10 37/416 (8.9%) 40
    PRURITUS 18/213 (8.5%) 28 52/416 (12.5%) 75
    RASH 17/213 (8%) 24 80/416 (19.2%) 116
    Vascular disorders
    HOT FLUSH 27/213 (12.7%) 27 24/416 (5.8%) 26
    HYPERTENSION 11/213 (5.2%) 16 33/416 (7.9%) 42

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

    Principal Investigators are NOT employed by the organization sponsoring the study.

    The Study being conducted under this Agreement is part of the Overall Study. Investigator is free to publish in reputable journals or to present at professional conferences the results of the Study, but only after the first publication or presentation that involves the Overall Study. The Sponsor may request that Confidential Information be deleted and/or the publication be postponed in order to protect the Sponsor's intellectual property rights.

    Results Point of Contact

    Name/Title Medical Communications
    Organization Hoffmann-La Roche
    Phone 800 821-8590
    Email genentech@druginfo.com
    Responsible Party:
    Hoffmann-La Roche
    ClinicalTrials.gov Identifier:
    NCT02340221
    Other Study ID Numbers:
    • GO29058
    • 2014-003185-25
    First Posted:
    Jan 16, 2015
    Last Update Posted:
    Jul 12, 2022
    Last Verified:
    Jul 1, 2022