Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT01120184
Other study ID # BO22589
Secondary ID 2009-017905-13
Status Completed
Phase Phase 3
First received
Last updated
Start date July 31, 2010
Est. completion date September 16, 2016

Study information

Verified date February 2024
Source Hoffmann-La Roche
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This randomized, 3-arm, multicenter, phase III study will evaluate the efficacy and safety of trastuzumab emtansine (T-DM1) with pertuzumab or trastuzumab emtansine (T-DM1) with pertuzumab-placebo (blinded for pertuzumab), versus the combination of trastuzumab (Herceptin) plus taxane (docetaxel or paclitaxel) in participants with HER2-positive progressive or recurrent locally advanced or previously untreated metastatic breast cancer. Participants will be randomized to 1 of 3 treatment arms (Arms A, B or C). Arm A will be open-label, whereas Arms B and C will be blinded.


Recruitment information / eligibility

Status Completed
Enrollment 1095
Est. completion date September 16, 2016
Est. primary completion date September 30, 2014
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - Adult participants >/=18 years of age - HER2-positive breast cancer - Histologically or cytologically confirmed adenocarcinoma of the breast with locally recurrent or metastatic disease, and be a candidate for chemotherapy. Participants with locally advanced disease must have recurrent or progressive disease, which must not be amenable to resection with curative intent. - Participants must have measurable and/or non-measurable disease which must be evaluable per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 - Eastern Cooperative Oncology Group (ECOG) Performance Status 0 or 1 - Adequate organ function as determined by laboratory results Exclusion Criteria: - History of prior (or any) chemotherapy for metastatic breast cancer or recurrent locally advanced disease - An interval of <6 months from the last dose of vinca-alkaloid or taxane cytotoxic chemotherapy until the time of metastatic diagnosis - Hormone therapy <7 days prior to randomization - Trastuzumab therapy and/or lapatinib (neo- or adjuvant setting) <21 days prior to randomization - Prior trastuzumab emtansine or pertuzumab therapy

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
docetaxel
75 mg/m2 or 100 mg/m2 intravenously every 3 weeks for a minimum of 6 cycles.
paclitaxel
80 mg/m2 intravenously weekly for a minimum of 18 weeks
pertuzumab
840 mg intravenously on day 1 of cycle 1 followed by 420 mg intravenously every 3 weeks in subsequent cycles
pertuzumab-placebo
840 mg intravenously on day 1 of cycle 1 followed by 420 mg intravenously every 3 weeks in subsequent cycles
trastuzumab [Herceptin]
trastuzumab [Herceptin] doses when administered with docetaxel: 8 mg/kg intravenously on cycle 1 followed by 6 mg/kg every 3 weeks in subsequent cycles or trastuzumab (Herceptin) doses when administered with paclitaxel: 4 mg/kg intravenously on day 1 of cycle 1 followed by 2 mg/kg weekly starting on day 8 of cycle 1.
trastuzumab emtansine
3.6 mg/kg intravenously every 3 weeks

Locations

Country Name City State
Argentina Fundación Investigar Buenos Aires
Argentina Centro Medico San Roque San Miguel de Tucuman
Australia Royal Adelaide Hospital; Oncology Adelaide South Australia
Australia Wesley Medical Centre; Clinic For Haematology and Oncology Auchenflower Queensland
Australia Peter Maccallum Cancer Institute; Medical Oncology Melbourne Victoria
Australia Mater Misericordiae Hospital; Chemotherapy Cottage Sydney New South Wales
Australia Calvary Mater Newcastle; Medical Oncology Waratah New South Wales
Austria Lkh Salzburg - Univ. Klinikum Salzburg; Iii. Medizinische Abt. Salzburg
Austria Medizinische Universität Wien; Univ.Klinik für Innere Medizin I - Abt. für Onkologie Wien
Bahamas Oncology Consultants Limited Nassau
Belgium Cliniques Universitaires St-Luc Bruxelles
Belgium Clinique Ste-Elisabeth Namur
Belgium Sint Augustinus Wilrijk Wilrijk
Bosnia and Herzegovina University Clinical Centre of the Republic of Srpska Banja Luka
Bosnia and Herzegovina Clinic of Oncology, University Clinical Center Sarajevo Sarajevo
Brazil Clinica de Neoplasias Litoral Itajai SC
Brazil Liga Norte Riograndense Contra O Câncer Natal RN
Brazil Clinica de Oncologia de Porto Alegre - CliniOnco Porto Alegre RS
Brazil Hospital Nossa Senhora da Conceicao Porto Alegre RS
Brazil Hospital Sao Lucas - PUCRS Porto Alegre RS
Brazil *X*Instituto Nacional do Cancer - INCA Rio de Janeiro RJ
Brazil Hospital Perola Byington Sao Paulo SP
Brazil Instituto do Cancer do Estado de Sao Paulo - ICESP Sao Paulo SP
Canada Cross Cancer Institute; Clinical Trials Edmonton Alberta
Canada CHU de Québec ? Hôpital du Saint-Sacrement / ONCOLOGY Quebec
Canada Cuse - Centre Universitaire De Sante; Site Fleurimont Sherbrooke Quebec
Canada North York General Hospital Toronto Ontario
Colombia Clinica del Country Bogota
Colombia Instituto Colombiano Para El Avance De La Medicina: Icamedic Bucaramanga
Colombia Centro Medico Imbanaco Cali
Colombia Instituto Cancerologico de Nariño Pasto
Czechia Masarykuv onkologicky ustav Brno
Czechia Fakultni nemocnice Olomouc; Onkologicka klinika Olomouc
Czechia Fakultni Nemocnice Na Bulovce; Ustav Radiacni Onkologie Praha
Czechia Fakultni Poliklinika Vseobecne Fakultni Niemocnice; Onkologicka Klinika Praha 2
Denmark Vejle Sygehus; Onkologisk Afdeling Vejle
France HOPITAL JEAN MINJOZ; Oncologie Besancon
France Centre Jean Perrin; Hopital De Jour Clermont Ferrand
France Centre Oscar Lambret; Senologie Lille
France Institut Paoli Calmettes; Oncologie Medicale Marseille
France Institut régional du Cancer Montpellier Montpellier
France Centre D'Oncologie de Gentilly; Oncology Nancy
France Hopital Saint Louis; Service Onco Thoracique Paris
France HOPITAL TENON; Cancerologie Medicale Paris
France Institut Curie; Oncologie Medicale Paris
France Chu La Miletrie; Radiotherapie Poitiers
France Centre Rene Huguenin; ONCOLOGIE GENETIQUE Saint Cloud
France Institut de Cancerologie de La Loire; Radiotherapie St Priest En Jarez
Germany Klinikum Essen-Mitte Ev. Huyssens-Stiftung / Knappschafts GmbH; Klinik für Senologie / Brustzentrum Essen
Germany Universitätsklinikum Halle (Saale); Universitätsklinik Und Poliklinik Für Gynäkologie Halle
Germany Facharztzentrum Eppendorf, Studien GbR Hamburg
Germany MVZ Onko Medical GmbH Hannover, Ralf Lohse (Geschäftsführer) Hannover
Germany Nationales Centrum für Tumorerkrankungen (NCT) ; Gyn. Onk. Frauenklinik; Uniklinikum Heidelberg Heidelberg
Germany Universitätsklinikum Magdeburg; Frauenheilkunde & Geburtshilfe Magdeburg
Germany Universitätsmedizin Mainz; Klinik u. Poliklinik f. Geburtshilfe u. Frauenheilkunde Mainz
Germany Mühlenkreiskliniken; Johannes Wesling Klinikum Minden; Klinik für Frauenheilkunde und Geburtshilfe Minden
Germany Rotkreuzklinikum München; Frauenklinik Muenchen
Germany Klinikum Mutterhaus der Borromaeerinnen gGmbH; Haematologie/Onkologie Trier
Greece Alexandras General Hospital of Athens; Oncology Department Athens
Greece Univ General Hosp Heraklion; Medical Oncology Heraklion
Greece University Hospital of Larissa; Oncology Larissa
Guatemala Centro Oncológico Sixtino / Centro Oncológico SA Guatemala
Guatemala Grupo Angeles Guatemala City
Hungary Semmelweis Egyetem Aok; Iii.Sz. Belgyogyaszati Klinika Budapest
Hungary Semmelweis Egyetem Onkologiai Központ Budapest
Hungary Szent Margit Hospital; Dept. of Oncology Budapest
Hungary Pécsi Tudományegyetem; Klinikai Központ Onkoterápiás Intézet Pécs
Italy Centro Catanese Di Oncologia; Oncologia Medica Catania Sicilia
Italy Campus Universitario S.Venuta; Centro Oncologico T.Campanella Catanzaro Calabria
Italy IRST Istituto Scientifico Romagnolo Per Lo Studio E Cura Dei Tumori, Sede Meldola; Oncologia Medica Meldola Emilia-Romagna
Italy Ospedale Papardo- Piemonte;Oncologia Medica Messina Sicilia
Italy Istituto Europeo Di Oncologia Milano Lombardia
Italy A.O. Universitaria Policlinico Di Modena; Ematologia Modena Emilia-Romagna
Italy Fondazione Salvatore Maugeri Pavia Lombardia
Italy Azienda Ospedaliera Di Perugia Ospedale s. Maria Della Misericordia; Oncologia Medica Perugia Umbria
Italy Ospedale Misericordia E Dolce; Oncologia Medica Prato Toscana
Italy Arcispedale Santa Maria Nuova; Oncologia Reggio Emilia Emilia-Romagna
Italy Ospedale Degli Infermi; Divisione Di Oncologia Rimini Emilia-Romagna
Italy IRCCS Istituto Clinico Humanitas; Oncologia Rozzano (MI) Lombardia
Italy Azienda Ospedaliero-Universitaria Dipartimento Interaziendale Di Oncologia Udine Friuli-Venezia Giulia
Japan Aichi Cancer Center Hospital, Breast Oncology Aichi
Japan Natl Hosp Org Shikoku; Cancer Ctr, Surgery Ehime
Japan National Hospital Organization Kyushu Cancer Center;Breast Oncology Fukuoka
Japan Gifu University Hospital; Digestive Surgery Gifu
Japan Hiroshima University Hospital; Breast Surgery Hiroshima
Japan National Hospital Organization Hokkaido Cancer Center; Breast Surgery Hokkaido
Japan Hyogo Cancer Center; Breast Surgery Hyogo
Japan Hyogo College Of Medicine; Breast And Endocrine Surgery Hyogo
Japan Kanazawa University Hospital; Breast Oncology Ishikawa
Japan Iwate Med Univ School of Med; Surgery Iwate
Japan Sagara Hospital; Breast Surgery Kagoshima
Japan Tokai University Hospital, Breast Surgery Kanagawa
Japan Kumamoto City Hospital, Breast and Endocrine Surgery Kumamoto
Japan Kumamoto University Hospital; Breast and Endocrine Surgery Kumamoto
Japan Kyoto University Hospital; Breast Surgery Kyoto
Japan Tohoku University Hospital; General Surgery Miyagi
Japan Niigata Cancer Ctr Hospital; Breast Surgery Niigata
Japan Kawasaki Medical School Hospital; Breast and Thyroid Surgery Okayama
Japan National Hospital Organization Osaka National Hospital; Breast Surgery Osaka
Japan Osaka University Hospital; Breast and Endocrine Surgery Osaka
Japan Saitama Cancer Center, Breast Oncology Saitama
Japan Saitama Medical University International Medical Center; Medical Oncology Saitama
Japan Shizuoka Cancer Center; Breast Surgery Shizuoka
Japan Shizuoka General Hospital; Breast Surgery Shizuoka
Japan National Cancer Center Hospital; Medical Oncology Tokyo
Japan The Cancer Inst. Hosp. of JFCR; Breast Oncology Center Tokyo
Japan Tokyo Medical Uni. Hospital; Breast Oncology Tokyo
Japan Toranomon Hospital; Breast and Endocrine Surgery Tokyo
Korea, Republic of Seoul National University Bundang Hospital; Hematology Medical Oncology Gyeonggi-do
Korea, Republic of Asan Medical Center, Uni Ulsan Collegemedicine; Dept.Internal Medicine / Divisionhematology/Oncology Seoul
Korea, Republic of Korea University Guro Hospital; Oncology Seoul
Korea, Republic of Samsung Medical Centre; Division of Hematology/Oncology Seoul
Korea, Republic of Seoul National Uni Hospital; Dept. of Internal Medicine/Hematology/Oncology Seoul
Malaysia Pantai Hospital Kuala Lumpur; Dept of Oncology & Radiotherapy Kuala Lumpur FED. Territory OF Kuala Lumpur
Malaysia Beacon International Specialist Centre Petaling Jaya, Selangor Selangor
Malaysia Sunway Medical Centre Selangor
Mexico Centro de Investigacion; Clinica Del Pacifico Acapulco Guerrero
Mexico Centenario Hospital Miguel Hidalgo Aguascalientes
Mexico Centro Universitario Contra El Cancer Monterrey Nuevo LEON
Mexico Oaxaca Site Management Organization Oaxaca de Juárez Oaxaca
Mexico Hospital Privado San Jose; Oncologia Obregon Sonora
Mexico Centro Oncológico Estatal; ISSSEMYM Oncología Toluca
New Zealand Auckland city hospital; Auckland Regional Cancer Centre and Blood Service Auckland
New Zealand Waikato Hospital; Regional Cancer Center Hamilton
North Macedonia Private Health Organization Acibadem Sistina Hospital Skopje
Panama The Panama Clinic Panama
Peru Hospital Nacional Carlos Alberto Seguin Escobedo-Essalud; Oncology & Haemathology Arequipa
Peru Hospital Nacional Edgardo Rebagliati Martins; Oncologia Lima
Peru Unidad de Investigacion Oncologia Clinica ? Piura; Unidad de Oncología Clínica Piura
Philippines Cebu Cancer Institute; Perpetual Succour Hospital Cebu City
Philippines Cardinal Santos Medical Center San Juan
Poland Centrum Onkologii;Im. Franciszka Lukaszczyka;Onkologii Bydgoszcz
Poland Medical University of Gdansk Gdansk
Poland Centrum Onkologii, Instytut, Klinika Chemioterapii; Oddzial Chemoterapii Krakow
Poland COZL Oddzial Onkologii Klinicznej z pododdzialem Chemioterapii Dziennej Lublin
Poland Cent.Onkologii-Instytut im. M. S-Curie, Klinika Now. Piersi i Chirurgii Rekon Warszawa
Portugal IPO do Porto; Servico de Oncologia Medica Porto
Romania Coltea Hospital; Oncology Bucharest
Romania Prof. Dr. I. Chiricuta Institute of Oncology Cluj Napoca
Romania Cluj Clinical County Hospital; Oncology Dept Cluj-Napoca
Russian Federation Ivanovo Regional Oncology Dispensary Ivanovo
Russian Federation Clinical Oncology Dispensary of Ministry of Health of Tatarstan Kazan Tatarstan
Russian Federation Moscow city oncology hospital; #62 of Moscow Healthcare Department Moscow Moskovskaja Oblast
Russian Federation Blokhin Cancer Research Center; Combined Treatment Moskva Moskovskaja Oblast
Russian Federation State Budget Institution of Healthcare of Stavropol region Pyatigorsk Oncology Dispensary Pyatigorsk Stavropol
Russian Federation Ryazan State Medical University Named after I.P.Pavlov Ryazan Rjazan
Russian Federation SBI of Healthcare Samara Regional Clinical Oncology Dispensary Samara
Russian Federation SBI of Healthcare of Stavropol region Stavropol Regional Clinical Oncology Dispensary Stavropol
Russian Federation Tula Regional Oncology Dispensary Tula
Russian Federation GUZ Vladimir Regional Clinical Oncological Dispensary Vladimir
Spain Hospital del Mar; Servicio de Oncologia Barcelona
Spain Hospital General Universitario de Elche; Servicio de Oncologia Elche Alicante
Spain Complejo Hospitalario de Jaen-Hospital Universitario Medico Quirurgico; Servicio de Oncologia Jaen
Spain Complejo Hospitalario Universitario A Coruña (CHUAC, Materno Infantil), Oncología La Coruña
Spain Hospital General Universitario Gregorio Marañon; Servicio de Oncologia Madrid
Spain Hospital Universitario 12 de Octubre; Servicio de Oncologia Madrid
Spain Hospital Universitario La Paz; Servicio de Oncologia Madrid
Spain Hospital Clinico Universitario Virgen de la Victoria; Servicio de Oncologia Malaga
Spain Hospital Univ Vall d'Hebron; Servicio de Oncologia Sant Andreu de La Barca Barcelona
Spain Complejo Hospitalario Universitario de Santiago (CHUS) ; Servicio de Oncologia Santiago de Compostela LA Coruña
Spain Hospital Clinico Universitario de Valencia; Servicio de Onco-hematologia Valencia
Sweden Skånes Universitetssjukhus; Kliniska Forskningsenheten Onkologimottagning medicinsk behandling Malmö
Switzerland Kantonsspital Baden; Medizinische Klinik, Onkologie Baden
Switzerland Universitaetsspital Basel; Onkologie Basel
Switzerland Kantonsspital Graubünden Medizin Onkologie; Onkologie und Hämatologie Chur
Taiwan Changhua Christian Hospital; Hematology-Oncology Changhua
Taiwan Kaohsiung Medical Uni Chung-Ho Hospital; Dept of Surgery Kaohsiung
Taiwan National Taiwan Uni Hospital; Dept of Oncology Taipei
Taiwan Tri-Service General Hospital; Hematology and Oncology Taipei
Thailand Faculty of Med. Siriraj Hosp.; Med.-Div. of Med. Oncology Bangkok
Thailand National Cancer Inst. Bangkok
Thailand Rajavithi Hospital; Division of Medical Oncology Bangkok
Thailand Ramathibodi Hospital; Dept of Med.-Div. of Med. Onc Bangkok
Thailand Maharaj Nakorn Hospital; Internal Medicine Chiang Mai
Thailand Songklanagarind Hospital; Department of Oncology Songkhla
Turkey Baskent University Adana Dr. Turgut Noyan Practice and Research Hospital; Medical Oncology Adana
Turkey Cukurova Uni Faculty of Medicine; Medical Oncology Adana
Turkey Gazi Uni Medical Faculty Hospital; Oncology Dept Ankara
Turkey Ege Uni Medical Faculty Hospital; Oncology Dept Izmir
United Kingdom Bristol Haematology and Oncology centre Bristol
United Kingdom Cambridge University Hospitals NHS Foundation Trust Cambridge
United Kingdom Royal Cornwall Hospital; Dept of Clinical Oncology Cornwall
United Kingdom The Beatson West of Scotland Cancer Centre; Cancer Clinical Trials Unit Glasgow
United Kingdom Leicester Royal Infirmary; Dept. of Medical Oncology Leicester
United Kingdom Guys Hospital; Management Offices London
United Kingdom Royal Free Hospital; Dept of Oncology London
United Kingdom Royal Marsden Hospital; Dept of Med-Onc London
United Kingdom Christie Hospital; Breast Cancer Research Office Manchester
United Kingdom Nottingham City Hospital; Oncology Nottingham
United Kingdom Peterborough City Hospital; Oncology Ward Peterborough
United Kingdom Queen Alexandra Hospital, Portsmouth Portsmouth
United Kingdom Weston Park Hospital; Cancer Clinical Trials Centre Sheffield
United Kingdom Southampton General Hospital; Somers Cancer Research Building Southampton
United Kingdom Uni Hospital of North Staffordshire; Staffordshire Oncology Centre Stoke-on-Trent
United Kingdom Royal Marsden Hospital; Dept of Medical Oncology Sutton
United States The Don & Sybil Harrington Cancer Center; Department of Clinical Research Amarillo Texas
United States Anne Arundel Health System Research Instit-Annapolis Oncology Ctr Annapolis Maryland
United States University Cancer & Blood Center, LLC; Research Athens Georgia
United States Innovative Medical Research of South Florida Aventura Florida
United States Mountain States Tumor Inst. Boise Idaho
United States Boston Medical Center Boston Massachusetts
United States Charleston Oncology, P .A Charleston South Carolina
United States Medical University of SC (MUSC) Charleston South Carolina
United States Carolinas Hem-Oncology Assoc Charlotte North Carolina
United States SCRI Tennessee Oncology Chattanooga Chattanooga Tennessee
United States Uni of Chicago Chicago Illinois
United States Oncology Hematology Care - SCRI Cincinnati Ohio
United States South Carolina Oncology Associates - SCRI Columbia South Carolina
United States Northwest Oncology/ Hematology Assoc. Coral Springs Florida
United States Uni of Texas Southwestern Medical Center Dallas Texas
United States Decatur Memorial Hospital Decatur Illinois
United States The Providence Regional Medical Center Everett Everett Washington
United States Sanford Roger Maris Cancer Center Fargo North Dakota
United States San Juan Oncology Associates Farmington New Mexico
United States Florida Cancer Specialists; SCRI Fort Myers Florida
United States Queens Medical Associates Fresh Meadows New York
United States West Clinic Germantown Tennessee
United States Spectrum Health Grand Rapids Grand Rapids Michigan
United States Hackensack Uni Medical Center; Northern Nj Cancer Center Hackensack New Jersey
United States Ingalls Memorial Hospital Harvey Illinois
United States Penn State Milton S. Hershey Medical Center Hershey Pennsylvania
United States Carolina Oncology Specialists, PA - Hickory Hickory North Carolina
United States Cancer Research Center of Hawaii; Clinical Sciences Honolulu Hawaii
United States Uni of Texas - Md Anderson Cancer Center; Dept of Breast Medical Oncology Houston Texas
United States Indiana University Melvin and Bren Simon Cancer Center Indianapolis Indiana
United States Mayo Clinic-Jacksonville Jacksonville Florida
United States Queens Hospital Center Jamaica New York
United States Scripps Cancer Center La Jolla California
United States University of California; Moores Cancer Center La Jolla California
United States Horizon Oncology Research, Inc. Lafayette Indiana
United States Arena Oncology Associates Lake Success New York
United States Dartmouth Hitchcock Med Center Lebanon New Hampshire
United States Uni of Arkansas For Medical Sciences; Arkansas Cancer Research Center Little Rock Arkansas
United States James Graham Brown Cancer Center, University of Louisville Louisville Kentucky
United States Loyola University Med Center Maywood Illinois
United States Uni of Miami School of Medicine; Sylvester Comprehensive Cancer Center Miami Florida
United States University of Minnesota. Minneapolis Minnesota
United States Clnc L Trials & Rsch Assoc-Inc Montebello California
United States Tennessee Onc., PLLC - SCRI Nashville Tennessee
United States Vanderbilt-Ingram Cancer Ctr Nashville Tennessee
United States Cancer Inst. of New Jersey New Brunswick New Jersey
United States Smilow Cancer Hospital at Yale New Haven New Haven Connecticut
United States Icahn School of Medicine at Mount Sinai New York New York
United States Weill Medical College of Cornell Uni New York New York
United States Northern Utah Associates Ogden Utah
United States Illinois Cancer Care Peoria Illinois
United States Mayo Clinic Rochester Rochester Minnesota
United States St. John'S Mercy Medical Center; David C. Pratt Cancer Center Saint Louis Missouri
United States Southern California Kaiser Permanente San Diego California
United States Breastlink Medical Group Inc Santa Ana California
United States New England Cancer Specialists Scarborough Maine
United States University of Washington Seattle Washington
United States Avera Cancer Institute Sioux Falls South Dakota
United States Mercy Clinic Cancer & Hematology Springfield Missouri
United States Northwest Medical Specialties Tacoma Washington
United States Kaiser Permanente; Oncology Clinical Trials Vallejo California
United States Marion L. Shepard Cancer Center Washington North Carolina

Sponsors (2)

Lead Sponsor Collaborator
Hoffmann-La Roche Genentech, Inc.

Countries where clinical trial is conducted

United States,  Argentina,  Australia,  Austria,  Bahamas,  Belgium,  Bosnia and Herzegovina,  Brazil,  Canada,  Colombia,  Czechia,  Denmark,  France,  Germany,  Greece,  Guatemala,  Hungary,  Italy,  Japan,  Korea, Republic of,  Malaysia,  Mexico,  New Zealand,  North Macedonia,  Panama,  Peru,  Philippines,  Poland,  Portugal,  Romania,  Russian Federation,  Spain,  Sweden,  Switzerland,  Taiwan,  Thailand,  Turkey,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Percentage of Participants With Death or Disease Progression According to Independent Review Facility (IRF) Assessment Tumor assessments were performed according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. Disease progression was defined as a greater than or equal to (=) 20 percent (%) and 5-millimeter (mm) increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). The percentage of participants with death or disease progression was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Primary Progression-Free Survival (PFS) According to IRF Assessment Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. PFS was defined as the time from randomization to first documented disease progression or death from any cause. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). Median duration of PFS was estimated using Kaplan-Meier analysis, and corresponding confidence intervals (CIs) were computed using the Brookmeyer-Crowley method. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants Who Died Prior to Clinical Cutoff The percentage of participants who died prior to clinical cutoff was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 70 months from randomization until clinical cutoff of 15-May-2016 (every 3 months until death, loss to follow-up, withdrawal, or study termination)
Secondary Overall Survival (OS) at Clinical Cutoff OS was defined as the time from randomization to death from any cause. Median duration of OS was estimated using Kaplan-Meier analysis, and corresponding CIs were computed using the Brookmeyer-Crowley method. Up to 70 months from randomization until clinical cutoff of 15-May-2016 (every 3 months until death, loss to follow-up, withdrawal, or study termination)
Secondary Percentage of Participants With Death or Disease Progression According to Investigator Assessment Tumor assessments were performed by the investigator according to RECIST version 1.1. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). The percentage of participants with death or disease progression was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary PFS According to Investigator Assessment Tumor assessments were performed by the investigator according to RECIST version 1.1. PFS was defined as the time from randomization to first documented disease progression or death from any cause. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). Median duration of PFS was estimated using Kaplan-Meier analysis, and corresponding CIs were computed using the Brookmeyer-Crowley method. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants Experiencing Treatment Failure Treatment failure was defined as the discontinuation of all study medications in the treatment arm for any reason including disease progression, treatment toxicity, death, physician decision, or participant withdrawal. The percentage of participants with treatment failure was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014
Secondary Time to Treatment Failure (TTF) Treatment failure was defined as the discontinuation of all study medications in the treatment arm for any reason including disease progression, treatment toxicity, death, physician decision, or participant withdrawal. TTF was defined as the time from randomization to treatment failure. Median TTF was estimated using Kaplan-Meier analysis, and corresponding CIs were computed using the Brookmeyer-Crowley method. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014
Secondary One-Year Survival Rate The percentage of participants alive at 1 year after randomization was estimated as the one-year survival rate using Kaplan-Meier analysis, and corresponding CIs were computed using Greenwood's estimate of the standard error. From randomization until 1 year
Secondary Percentage of Participants With Grade =3 Adverse Events Adverse events were graded according to National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Grade 3: Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self care activity of daily living with inability to perform bathing, dressing and undressing, feeding self, using the toilet, taking medications but not bedridden. Grade 4: An immediate threat to life. Urgent medical intervention is required in order to maintain survival. Grade 5: Death. Up to 50 months from randomization until clinical cutoff of 16-Sept-2014 (continuously until 28 days after last dose
Secondary Percentage of Participants Who Died at 2 Years From randomization until 2 years
Secondary Overall Survival Truncated at 2 Years Overall Survival truncated at 2 years was defined as the percentage of participants alive at 2 years. From randomization until 2 years
Secondary Percentage of Participants With Grade 5 Adverse Events Adverse events were graded according to NCI CTCAE version 4.0. Grade 5 adverse events are those events which led to death. Up to 50 months from randomization until clinical cutoff of 16-Sept-2014 (continuously until 28 days after last dose)
Secondary Percentage of Participants With Grade 3-4 Laboratory Parameters Laboratory results were graded according to NCI CTCAE version 4.0. Grade 3: Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self care activity of daily living with inability to perform bathing, dressing and undressing, feeding self, using the toilet, taking medications but not bedridden. Grade 4: An immediate threat to life. Urgent medical intervention is required in order to maintain survival. Day 1, 8, and 15 of Cycle 1-3 and on Day 1 of each subsequent cycle up to 50 months from randomization until clinical cutoff of 16-Sept-2014
Secondary Percentage of Participants With Decline of =2 Points From Baseline in Eastern Cooperative Oncology Group (ECOG) Performance Status The ECOG performance status is a scale used to quantify cancer participants' general well-being and activities of daily life. The scale ranges from 0 to 5, with 0 denoting perfect health and 5 indicating death. The 6 categories are 0=Asymptomatic (Fully active, able to carry on all predisease activities without restriction), 1=Symptomatic but completely ambulatory (Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature), 2=Symptomatic, < 50% in bed during the day (Ambulatory and capable of all self-care but unable to carry out any work activities. Up and about more than 50% of waking hours), 3=Symptomatic, > 50% in bed, but not bedbound (Capable of only limited self-care, confined to bed or chair 50% or more of waking hours), 4=Bedbound (Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair), 5=Death. Baseline, Day 1 of every Cycle up to Clinical Data Cut (up to 48 months)
Secondary Hospitalization Days Hospitalization was defined as a non-administration-related hospitalization due to serious adverse event, while on study treatment. Reported values represent number of days admitted per participants. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014
Secondary Percentage of Participants With Hospitalization Hospitalization was defined as a non-administration-related hospitalization due to serious adverse event, while on study treatment. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014
Secondary Percentage of Participants With Objective Response According to IRF Assessment Objective response was defined as having complete response (CR) or partial response (PR), assessed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. CR was defined as the disappearance of all target and non-target lesions and short-axis reduction in pathological lymph nodes to <10 mm. PR was defined as a =30% decrease in sum of diameters of target lesions, taking as reference the Baseline sum. Response was determined using 2 consecutive tumor assessments at least 4 weeks apart. The percentage of participants with a best overall response of CR or PR (ie, the objective response rate [ORR]) was calculated as [number of participants meeting the respective criteria divided by the number analyzed] multiplied by 100. Corresponding CIs were computed using the Blyth-Still-Casella exact method. Up to 46 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants With Objective Response According to Investigator Assessment Objective response was defined as having CR or PR, assessed according to RECIST version 1.1, by investigator. CR was defined as the disappearance of all target and non-target lesions and short-axis reduction in pathological lymph nodes to <10 mm. PR was defined as a =30% decrease in sum of diameters of target lesions, taking as reference the Baseline sum. Response was determined using 2 consecutive tumor assessments at least 4 weeks apart. The percentage of participants with a best overall response of CR or PR (ie, the ORR) was calculated as [number of participants meeting the respective criteria divided by the number analyzed] multiplied by 100. Corresponding CIs were computed using the Blyth-Still-Casella exact method. Up to 46 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Duration of Response According to IRF Assessment Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. Duration of response was defined as the time from confirmed PR or CR to first documented disease progression or death from any cause. CR was defined as the disappearance of all target lesions and non-target lesions and short-axis reduction in pathological lymph nodes to <10 mm. PR was defined as a =30% decrease in sum of diameters of target lesions, taking as reference the Baseline sum. Response was determined using 2 consecutive tumor assessments at least 4 weeks apart. Median duration of response was estimated using Kaplan-Meier analysis, and corresponding CIs were computed using the Brookmeyer-Crowley method. Up to 46 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants With a Best Overall Response of CR, PR, or Stable Disease (SD) According to IRF Assessment Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. CR was defined as the disappearance of all target and non-target lesions and short-axis reduction in pathological lymph nodes to <10 mm. PR was defined as a =30% decrease in sum of diameters of target lesions, taking as reference the Baseline sum. SD was defined as neither sufficient shrinkage to quality for PR nor sufficient (20%) increase to qualify for disease progression. Response was determined using 2 consecutive tumor assessments at least 4 weeks apart. The percentage of participants with a best overall response of CR, PR, or SD was calculated as [number of participants meeting the respective criteria divided by the number analyzed] multiplied by 100. Up to 46 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants Experiencing a Clinically Significant Increase in Taxane-Related Treatment Symptoms as Measured by Taxane Subscale of the Functional Assessment of Cancer Therapy (FACT) Taxane (FACT-TaxS) Score The FACT-Taxane is a self-reported instrument which measures the health-related quality of life (HRQOL) of participants receiving taxane-containing chemotherapy. The FACT-TaxS consists of 16 items including 11 neurotoxicity-related questions and 5 additional questions assessing arthralgia, myalgia, and skin discoloration. Items are rated from 0 (not at all) to 4 (very much) and a total score is inversely derived. Scores may range from 0 to 64, with higher scores indicating fewer/no symptoms. A minimally clinically important difference in treatment-related symptoms was defined as a =5% decrease (ie, 3.2 points) in FACT-TaxS score from Baseline. The percentage of participants with treatment-related symptoms was calculated using following formula: [number of participants meeting the above threshold divided by the number analyzed] multiplied by 100. Corresponding CIs were computed using the Blyth-Still-Casella exact method. Up to 39 months from randomization until clinical cutoff of 16-Sept-2014 (at Baseline, on Day 1 of Cycles 2 to 8 and every even-numbered cycle thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants Reporting Nausea According to the Relevant Single Items of The FACT Colorectal Cancel (FACT-C) Module The FACT-C is a self-reported instrument which measures HRQOL pertaining to colorectal cancer. Response options on each question may range from 0 (not at all) to 4 (very much). The percentage of participants with nausea was calculated using following formula: [number of participants with any level of either symptom divided by the number analyzed] multiplied by 100. At Baseline, Day 8 of Cycle 1, and Days 1 and 8 of Cycle 2
Secondary Percentage of Participants Reporting Diarrhea According to the Relevant Single Items of The FACT-C Module The FACT-C is a self-reported instrument which measures HRQOL pertaining to colorectal cancer. Response options on each question may range from 0 (not at all) to 4 (very much). The percentage of participants with diarrhea was calculated using following formula: [number of participants with any level of either symptom divided by the number analyzed] multiplied by 100. At Baseline, Day 8 of Cycle 1, and Days 1 and 8 of Cycle 2
Secondary Percentage of Participants With a Clinically Significant Deterioration in Health Related Quality of Life (HRQoL) as Measured by FACT Breast (FACT-B) Trial Outcome Index-Physical Function Breast (TOI-PFB) Score The FACT-B is a self-reported instrument which measures HRQOL of participants with breast cancer. It consists of 5 subscales including physical well-being (PWB), social well-being (SWB), emotional well-being (EWB), functional well-being (FWB), and a breast cancer subscale (BCS). The TOI-PFB score is taken by adding the scores from the PWB (7 items), FWB (7 items), and BCS (9 items) subscales. Items are rated from 0 (not at all) to 4 (very much) and a total score is derived. Scores may range from 0 to 92, with higher scores indicating better HRQOL. A 5 point change has been identified as the clinically minimal important difference (CMID) on the FACT-TOI-PFB scale. The percentage of participants with deterioration was calculated as [number of participants meeting the above threshold divided by the number analyzed] multiplied by 100. Up to 39 months from randomization until clinical cutoff of 16-Sept-2014 (at Baseline, on Day 1 of Cycles 2 to 8 and every even-numbered cycle thereafter and/or up to 42 days after last dose)
Secondary Time to Deterioration in HRQoL as Assessed by FACT-B TOI-PFB Score The FACT-B is a self-reported instrument which measures HRQOL of participants with breast cancer. It consists of 5 subscales including PWB, SWB, EWB, FWB, and BCS. The TOI-PFB score is taken by adding the scores from the PWB (7 items), FWB (7 items), and BCS (9 items) subscales. Items are rated from 0 (not at all) to 4 (very much) and a total score is derived. Scores may range from 0 to 92, with higher scores indicating better HRQOL. A 5 point change has been identified as the clinically minimal important difference (CMID) on the FACT-TOI-PFB scale. Time to deterioration was defined as the time from Baseline until the first decrease in FACT-B TOI-PFB score. Median time to deterioration was estimated using Kaplan-Meier analysis, and corresponding CIs were computed using the Brookmeyer-Crowley method. Baseline up to 39 months from randomization until clinical cutoff of 16-Sept-2014
Secondary Change From Baseline in Rotterdam Symptom Checklist (RSCL) Activity Level Scale Score The RSCL is a self-reported instrument which consists of 4 domains including physical symptom distress, psychological distress, activity level, and overall global life quality. Only the activity level scale was collected and assessed. Scores may range from 0 to 100, with higher scores indicating increased burden of disease. Mean RSCL activity scale score changes were calculated as [mean score at the assessment visit minus mean score at Baseline]. The higher the score, the higher the level of impairment or burden. Baseline, Cycle 7 (Week 18)
Secondary Change From Baseline in Work Productivity According to Work Productivity and Activity Impairment (WPAI) Questionnaire Score The WPAI is a patient-reported measure which assesses the effect of general health and symptom severity on work productivity and regular activities. The General Health questionnaire asks participants to estimate the number of hours missed from work due to reasons related and unrelated to their health problems, as well as the total number of hours actually worked in the preceding 7-day period. The percentage of participants reporting that they were employed (working for pay) was assessed at baseline was assessed along with Absenteeism (work time missed), Presenteeism (impairment at work / reduced on-the-job effectiveness), Work productivity loss (overall work impairment / absenteeism plus presenteeism), and Activity Impairment. The reported changes represent change from baseline at Cycle 7. The score range for the scales of the WPAI is between 0 (no effect) to 100% (max effect) Baseline, Cycle 7 (Week 18)
Secondary Change From Baseline in Activity Impairment According to Work Productivity and Activity Impairment (WPAI) Questionnaire Score The WPAI is a patient-reported measure which assesses the effect of general health and symptom severity on work productivity and regular activities. The General Health questionnaire asks participants to estimate the number of hours missed from work due to reasons related and unrelated to their health problems, as well as the total number of hours actually worked in the preceding 7-day period. The percentage of participants reporting that they were employed (working for pay) was assessed at baseline was assessed along with Absenteeism (work time missed), Presenteeism (impairment at work / reduced on-the-job effectiveness), Work productivity loss (overall work impairment / absenteeism plus presenteeism), and Activity Impairment. The reported changes represent change from baseline at Cycle 7. The score range for the scales of the WPAI is between 0 (no effect) to 100% (max effect). Baseline, Cycle 7 (Week 18)
Secondary Percentage of Participants With a Best Overall Response of CR or PR According to IRF Assessment Among Those With High Human Epidermal Growth Factor Receptor 2 (HER2) Messenger Ribonucleic Acid (mRNA) Levels Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. CR was defined as the disappearance of all target and non-target lesions and short-axis reduction in pathological lymph nodes to <10 mm. PR was defined as a =30% decrease in sum of diameters of target lesions, taking as reference the Baseline sum. Response was determined using 2 consecutive tumor assessments at least 4 weeks apart. The percentage of participants with a best overall response of CR or PR (ie, the ORR) was calculated as [number of participants meeting the respective criteria divided by the number analyzed] multiplied by 100. Up to 46 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants With a Best Overall Response of CR or PR According to IRF Assessment Among Those With Low HER2 mRNA Levels Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. CR was defined as the disappearance of all target and non-target lesions and short-axis reduction in pathological lymph nodes to <10 mm. PR was defined as a =30% decrease in sum of diameters of target lesions, taking as reference the Baseline sum. Response was determined using 2 consecutive tumor assessments at least 4 weeks apart. The percentage of participants with a best overall response of CR or PR (ie, the ORR) was calculated as [number of participants meeting the respective criteria divided by the number analyzed] multiplied by 100. Up to 46 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants With Death or Disease Progression According to IRF Assessment Among Those With High HER2 mRNA Levels Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). The percentage of participants with death or disease progression was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary PFS According to IRF Assessment Among Those With High HER2 mRNA Levels Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. PFS was defined as the time from randomization to first documented disease progression or death from any cause. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). Median duration of PFS was estimated using Kaplan-Meier analysis. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants With Death or Disease Progression According to IRF Assessment Among Those With Low HER2 mRNA Levels Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). The percentage of participants with death or disease progression was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary PFS According to IRF Assessment Among Those With Low HER2 mRNA Levels Tumor assessments were performed according to RECIST version 1.1, using radiographic images submitted to the IRF up to and including the confirmatory tumor assessment 4 to 6 weeks after study drug discontinuation. PFS was defined as the time from randomization to first documented disease progression or death from any cause. Disease progression was defined as a =20% and 5-mm increase in sum of diameters of target lesions, taking as reference the smallest sum obtained during the study, or appearance of new lesion(s). Median duration of PFS was estimated using Kaplan-Meier analysis. Up to 48 months from randomization until clinical cutoff of 16-Sept-2014 (at Screening, every 9 weeks for 81 weeks, then every 12 weeks thereafter and/or up to 42 days after last dose)
Secondary Percentage of Participants Who Died Prior to Clinical Cutoff Among Those With High HER2 mRNA Levels The percentage of participants who died prior to clinical cutoff was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 70 months from randomization until clinical cutoff of 15-May-2016 (every 3 months until death, loss to follow-up, withdrawal, or study termination)
Secondary OS at Clinical Cutoff Among Those With High HER2 mRNA Levels OS was defined as the time from randomization to death from any cause. Median duration of OS was estimated using Kaplan-Meier analysis. Up to 70 months from randomization until clinical cutoff of 15-May-2016 (every 3 months until death, loss to follow-up, withdrawal, or study termination)
Secondary Percentage of Participants Who Died Prior to Clinical Cutoff Among Those With Low HER2 mRNA Levels The percentage of participants who died prior to clinical cutoff was calculated as [number of participants with event divided by the number analyzed] multiplied by 100. Up to 70 months from randomization until clinical cutoff of 15-May-2016 (every 3 months until death, loss to follow-up, withdrawal, or study termination)
Secondary OS at Clinical Cutoff Among Those With Low HER2 mRNA Levels OS was defined as the time from randomization to death from any cause. Median duration of OS was estimated using Kaplan-Meier analysis. Reported upper bound of confidence interval for "Trastuzumab Emtansine + Placebo" and confidence interval values for "Trastuzumab + Taxane" and "Trastuzumab Emtansine + Pertuzumab" are censored values. Up to 70 months from randomization until clinical cutoff of 15-May-2016 (every 3 months until death, loss to follow-up, withdrawal, or study termination)
See also
  Status Clinical Trial Phase
Recruiting NCT04681911 - Inetetamab Combined With Pyrotinib and Chemotherapy in the Treatment of HER2 Positive Metastatic Breast Cancer Phase 2
Terminated NCT04066790 - Pyrotinib or Trastuzumab Plus Nab-paclitaxel as Neoadjuvant Therapy in HER2-positive Breast Cancer Phase 2
Completed NCT04890327 - Web-based Family History Tool N/A
Completed NCT03591848 - Pilot Study of a Web-based Decision Aid for Young Women With Breast Cancer, During the Proposal for Preservation of Fertility N/A
Recruiting NCT03954197 - Evaluation of Priming Before in Vitro Maturation for Fertility Preservation in Breast Cancer Patients N/A
Terminated NCT02202746 - A Study to Assess the Safety and Efficacy of the VEGFR-FGFR-PDGFR Inhibitor, Lucitanib, Given to Patients With Metastatic Breast Cancer Phase 2
Active, not recruiting NCT01472094 - The Hurria Older PatiEnts (HOPE) With Breast Cancer Study
Recruiting NCT06057636 - Hypnosis for Pain in Black Women With Advanced Breast Cancer: A Feasibility Study N/A
Recruiting NCT06049446 - Combining CEM and Magnetic Seed Localization of Non-Palpable Breast Tumors
Recruiting NCT05560334 - A Single-Arm, Open, Exploratory Clinical Study of Pemigatinib in the Treatment of HER2-negative Advanced Breast Cancer Patients With FGFR Alterations Phase 2
Active, not recruiting NCT05501769 - ARV-471 in Combination With Everolimus for the Treatment of Advanced or Metastatic ER+, HER2- Breast Cancer Phase 1
Recruiting NCT04631835 - Phase I Study of the HS-10352 in Patients With Advanced Breast Cancer Phase 1
Completed NCT04307407 - Exercise in Breast Cancer Survivors N/A
Recruiting NCT03544762 - Correlation of 16α-[18F]Fluoro-17β-estradiol PET Imaging With ESR1 Mutation Phase 3
Terminated NCT02482389 - Study of Preoperative Boost Radiotherapy N/A
Enrolling by invitation NCT00068003 - Harvesting Cells for Experimental Cancer Treatments
Completed NCT00226967 - Stress, Diurnal Cortisol, and Breast Cancer Survival
Recruiting NCT06037954 - A Study of Mental Health Care in People With Cancer N/A
Recruiting NCT06006390 - CEA Targeting Chimeric Antigen Receptor T Lymphocytes (CAR-T) in the Treatment of CEA Positive Advanced Solid Tumors Phase 1/Phase 2
Recruiting NCT06019325 - Rhomboid Intercostal Plane Block on Chronic Pain Incidence and Acute Pain Scores After Mastectomy N/A