A Study to Compare Subcutaneous (SC) Versus Intravenous (IV) Administration of Herceptin (Trastuzumab) in Women With Human Epidermal Growth Factor Receptor (HER) 2-Positive Early Breast Cancer

December 21, 2017 updated by: Hoffmann-La Roche

A Phase III, Randomized Open-Label Study to Compare the Pharmacokinetics, Efficacy, and Safety of Subcutaneous (SC) Trastuzumab With Intravenous (IV) Trastuzumab Administered in Women With HER2-Positive Early Breast Cancer (EBC)

In this open-label multicenter trial, participants with operable or locally advanced breast cancer will be randomized to pre-operative treatment with 8 cycles of chemotherapy (4 cycles of docetaxel followed by 4 cycles of 5-fluorouracil, epirubicin, and cyclophosphamide) concurrent with either SC Herceptin or IV Herceptin. After surgery, participants will receive a further 10 cycles of SC or IV Herceptin as per randomization to complete 1 year of treatment. All cycles will be 21 days in length. After the end of study treatment, participants will be followed for safety and efficacy for up to 5 years or until disease recurrence, whichever is earlier.

Study Overview

Study Type

Interventional

Enrollment (Actual)

596

Phase

  • Phase 3

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Tucuman, Argentina, T4000IAK
        • Centro Medico San Roque; Oncology Dept
      • Tucuman, Argentina, T400IAK
        • Caipo; Oncology
    • BA
      • Salvador, BA, Brazil, 41253-190
        • Hospital Sao Rafael - HSR
    • PR
      • Curitiba, PR, Brazil, 80060-900
        • Hospital das Clinicas - UFPR; Quimioterapia
    • RN
      • Natal, RN, Brazil, 59040150
        • Liga Norte Riograndense Contra O Cancer
    • SC
      • Itajai, SC, Brazil, 88301-220
        • Clinica de Neoplasias Litoral
    • SP
      • Jau, SP, Brazil, 17210-080
        • Hospital Amaral Carvalho
      • Sao Paulo, SP, Brazil, 01246-000
        • Instituto do Cancer do Estado de Sao Paulo - ICESP
      • Sao Paulo, SP, Brazil, 01317-000
        • Hospital Perola Byington
      • Sorocaba, SP, Brazil, 18030-245
        • Instituto de Oncologia de Sorocaba - CEPOS
      • Quebec, Canada, G1S 4L8
        • CHU de Québec - Hôpital du Saint-Sacrement / ONCOLOGY
    • Quebec
      • Montreal, Quebec, Canada, H1T 2M4
        • Hopital Maisonneuve- Rosemont; Oncology
      • Bogota, Colombia, 000472
        • Hospital Universitario San Ignacio
      • Bogota, Colombia
        • Grupo Salud Coop
      • Pereira, Colombia, 600004
        • Oncólogos de Occidente
      • Olomouc, Czechia, 779 00
        • Fakultni nemocnice Olomouc; Onkologicka klinika
      • Pardubice, Czechia, 532 03
        • Krajska Nemocnice Pardubice Neurologicka Klinika
      • Praha, Czechia, 140 59
        • Fakultni Thomayerova Nemocnice; Onkologicke Oddeleni
      • Praha, Czechia, 180 81
        • Lekarske Fakulty Univerzity Karlovy Fakultni Nemocnice Na Bulovce; Ustav Radiacni Onkologie
      • Tallinn, Estonia, 13419
        • North Estonia Medical Centre Foundation; Oncology Centre
      • Tartu, Estonia, 50406
        • Tartu University Hospital; Clinic of Hematology and Oncology
      • Besancon, France, 25030
        • HOPITAL JEAN MINJOZ; Oncologie
      • Grenoble, France, 38000
        • Institut Daniel Hollard; Chimiotherapie Ambulatoire
      • La Tronche, France, 38700
        • Hopital Albert Michallon; Oncologie
      • Le Mans, France, 72015
        • Centre Jean Bernard
      • Paris, France, 75475
        • Hopital Saint Louis ; Service d Oncologie Medicale Fougere 6 (Pr Misset)
      • Reims, France, 51056
        • Institut Jean Godinot; Hopital De Jour
      • St Cloud, France, 92210
        • Centre Rene Huguenin; Medecine B
      • Berlin, Germany, 10719
        • Praxis Dr. Schoenegg
      • Berlin, Germany, 10117
        • CAMPUS CHARITÉ MITTE; Tagesklinik für Onkologie u.Hämatologie
      • Bonn, Germany, 53113
        • Johanniter GmbH; Johanniter-Krankenhaus; Internistische Onkologie
      • Dortmund, Germany, 44137
        • St. Johannes Hospital
      • Hannover, Germany, 30177
        • Gynaekologisch-Onkologische Schwerpunktpraxis Prof. Dr. med. Lueck, Dr. Schrader und Dr. Noeding
      • Leipzig, Germany, 04277
        • Sankt Elisabeth Krankenhaus; Gynaekology
      • Lemgo, Germany, 32657
        • Klinik Lippe Lemgo; Frauenklinik
      • Offenbach, Germany, 63069
        • Sana Klinikum Offenbach GmbH; Klinik für Gynäkologie & Geburtshilfe
      • Rodewisch, Germany, 08228
        • Klinik Obergöltzsch; Abt. Gynäkologie
      • Tübingen, Germany, 72076
        • Universitätsklinik Tübingen; Frauenklinik
      • Guatemala, Guatemala, 01010
        • Centro Oncologico S.A.
      • Hong Kong, Hong Kong, 852
        • Queen Mary Hospital; Surgery
      • Budapest, Hungary, 1083
        • Semmelweis Egyetem Onkologiai Központ
      • Gyor, Hungary, 9023
        • Hospital of Aladar Petz; Dept of Oncoradiology
      • Szeged, Hungary, 6720
        • Szegedi Tudomanyegyetem, AOK, Szent-Gyorgyi Albert Klinikai Kozpont, Onkoterapias Klinika
      • Jerusalem, Israel, 9112001
        • Hadassah Ein Karem Hospital; Oncology Dept
      • Petach Tikva, Israel, 49100
        • Rabin Medical Center; Oncology Dept
    • Campania
      • Napoli, Campania, Italy, 80131
        • Istituto Nazionale Tumori Irccs Fondazione g. Pascale;s.c. Ematologia Oncologica
    • Lombardia
      • Cremona, Lombardia, Italy, 26100
        • ASST DI CREMONA; Dip. Medicina - S.C. Oncologia
      • Pavia, Lombardia, Italy, 27100
        • Fondazione Salvatore Maugeri; Servizio Di Prevenzione Oncologica
      • Pavia, Lombardia, Italy, 27100
        • Fondazione Salvatore Maugeri
      • Incheon, Korea, Republic of, 405-760
        • Gachon Medical School Gil Medical Center; Medical Oncology
      • Seoul, Korea, Republic of, 136-705
        • Korea University Anam Hospital; Oncology Haemotology
      • Seoul, Korea, Republic of, 135-710
        • Samsung Medical Centre; Division of Hematology/Oncology
      • Seoul, Korea, Republic of, 138-736
        • Asan Medical Center, Uni Ulsan Collegemedicine; Dept.Internal Medicine / Divisionhematology/Oncology
      • Obregon, Mexico, 85000
        • Hospital Privado San Jose; Oncologia
      • Toluca, Mexico, 50180
        • Centro Oncológico Estatal; ISSSEMYM Oncología
      • Panama, Panama, 0834-02723
        • Centro Oncologico America
      • Arequipa, Peru, 04001
        • Hospital Nacional Carlos Alberto Seguin Escobedo-Essalud; Oncology & Haemathology
      • Lima, Peru, 41
        • Oncosalud Sac; Oncología
      • Lima, Peru, 11
        • Hospital Nacional Edgardo Rebagliati Martins; Oncologia
      • Piura, Peru, 20011
        • Unidad de Investigacion Oncologia Clinica - Piura; Unidad de Oncología Clínica
      • San Isidro, Peru, Lima 27
        • Clinica Ricardo Palma
      • Elblag, Poland, 82-300
        • Wojewodzki Szpital Zespolony; Oddział Onkologii
      • Lublin, Poland, 20-090
        • COZL Oddzial Onkologii Klinicznej z pododdzialem Chemioterapii Dziennej
      • Olsztyn, Poland, 10-513
        • Olsztyński Ośrodek Onkologiczny Kopernik sp. z o.o.
      • Warszawa, Poland, 02-781
        • Centrum Onkologii - Instytut im. Marii Skłodowskiej-Curie Klinika Nowotworów Piersi i Chirurgii
      • Ivanovo, Russian Federation, 153040
        • Ivanovo Regional Oncology Dispensary
      • Moscow, Russian Federation, 143423
        • Moscow city oncology hospital #62 of Moscow Healthcare Department
      • Moscow, Russian Federation, 115478
        • Blokhin Cancer Research Center; Combined Treatment
      • Pyatigorsk, Russian Federation, 357502
        • State Budget Institution of Healthcare of Stavropol region Pyatigorsk Oncology Dispensary
      • Ryazan, Russian Federation, 390011
        • SBI for HPE "Ryazan State Medical University n.a. I.P. Pavlov" of MoH of RF
      • Samara, Russian Federation, 443031
        • SBI of Healthcare Samara Regional Clinical Oncology Dispensary
      • Saratov, Russian Federation, 410053
        • Saratov Regional Clinical Hospital & Pathology Centre
      • St Petersburg, Russian Federation, 197022
        • Saint-Petersburg City Clinical Oncology Dispensary
      • Stavropol, Russian Federation, 355045
        • SBI of Healthcare of Stavropol region Stavropol Regional Clinical Oncology Dispensary
      • Tula, Russian Federation, 300053
        • Tula Regional Oncology Dispensary
      • Vladimir, Russian Federation, 600009
        • GUZ Vladimir Regional Clinical Oncological Dispensary
    • Leningrad
      • St Petersburg, Leningrad, Russian Federation, 197758
        • FSBI"National Medical Research Center of Oncology named after N.N.Petrov" MHRF
      • Kosice, Slovakia, 04001
        • Východoslovenský Onkologický Ústav
      • Poprad, Slovakia, 05801
        • Nzz - Oncology Outpatient Clinic
      • Bloemfontein, South Africa, 9301
        • National Hospital; Oncotherapy Dept
      • Cape Town, South Africa, 7506
        • Groote Schuur Hospital ( Uni of Capetown ); Oncology Dept
      • Cape Town, South Africa, 7506
        • Cancercare
      • Parktown, Johannesburg, South Africa, 2193
        • Wits Donald Gordon Clinical Trial Centre; Medical Oncology
      • Pretoria, South Africa, 0081
        • Pretoria-East Hospital; 1 Sanwood Park
      • Rondebosch, South Africa, 7700
        • Rondebosch Oncology Centre
      • Sandton, South Africa, 2196
        • Sandton Oncology Centre
      • Madrid, Spain, 28007
        • Hospital General Universitario Gregorio Marañon; Servicio de Oncologia
      • Valencia, Spain, 46010
        • Hospital Clinico Universitario de Valencia; Servicio de Onco-hematologia
      • Zaragoza, Spain, 50009
        • Hospital Universitario Miguel Servet; Servicio Oncologia
    • LA Coruña
      • Santiago de Compostela, LA Coruña, Spain, 15706
        • Complejo Hospitalario Universitario de Santiago (CHUS) ; Servicio de Oncologia
    • Tarragona
      • Reus, Tarragona, Spain, 43204
        • Hospital Universitari Sant Joan de Reus; Servicio de Oncologia
      • Lund, Sweden, 22185
        • Skånes University Hospital, Skånes Department of Onclology
      • Uppsala, Sweden, 75185
        • Akademiska sjukhuset, Onkologkliniken
      • Changhua, Taiwan, 500
        • Changhua Christian Hospital; Dept of Surgery
      • Taipei, Taiwan, 112
        • Koo Foundation Sun Yat-Sen Cancer Center; Hemato-Oncology
      • Taipei City, Taiwan, 112
        • Taipei Veterans General Hospital
      • Taoyuan, Taiwan, 333
        • Chang Gung Medical Foundation-Taipei
      • Bangkok, Thailand, 10330
        • Chulalongkorn Hospital; Medical Oncology
      • Bangkok, Thailand, 10700
        • Faculty of Med. Siriraj Hosp.; Med.-Div. of Med. Oncology
      • Bangkok, Thailand, 10400
        • National Cancer Inst.
      • Bangkok, Thailand, 10400
        • Phramongkutklao Hospital;Dept Surgery/Surgical Oncology Unit
      • Songkhla, Thailand, 90110
        • Prince of Songkla Uni ; Unit of Medical Oncology
      • Antalya, Turkey, 07070
        • Akdeniz University Medical Faculty; Medical Oncology Department
      • Istanbul, Turkey, 34390
        • Istanbul Uni of Medicine Faculty; Oncology Dept
      • Izmir, Turkey, 35340
        • Dokuz Eylul Uni Medical Faculty; Oncology Dept
      • Sıhhiye, Ankara, Turkey, 06100
        • Hacettepe Uni Medical Faculty Hospital; Oncology Dept

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (ADULT, OLDER_ADULT)

Accepts Healthy Volunteers

No

Genders Eligible for Study

Female

Description

Inclusion Criteria:

  • Adult women greater than or equal to (≥) 18 years of age
  • Non-metastatic primary invasive adenocarcinoma of the breast clinical stage I to IIIC, including inflammatory and multicentric/multifocal breast cancer, with tumor size ≥1 centimeter (cm) by ultrasound or ≥2 cm by palpation, centrally confirmed HER2-positive (immunohistochemical score [IHC] 3+ or in situ hybridization [ISH]-positive)
  • At least 1 measurable lesion in breast or lymph nodes (≥1 cm by ultrasound or ≥2 cm by palpation), except for inflammatory carcinoma (T4d)
  • Baseline left ventricular ejection fraction (LVEF) ≥55%
  • Eastern Cooperative Oncology Group (ECOG) status of 0 or 1
  • Adequate organ function at Baseline

Exclusion Criteria:

  • History of any prior (ipsilateral and/or contralateral) invasive breast carcinoma
  • Past or current history of malignant neoplasms, except for curatively treated basal and squamous cell carcinoma of the skin and in situ carcinoma of the cervix
  • Metastatic disease
  • Any prior therapy with anthracyclines
  • Prior anti-HER2 therapy or biologic or immunotherapy
  • Serious cardiac illness
  • Pregnant or lactating women

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: TREATMENT
  • Allocation: RANDOMIZED
  • Interventional Model: PARALLEL
  • Masking: NONE

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
ACTIVE_COMPARATOR: Herceptin IV + Chemotherapy
Participants will receive Herceptin via IV infusion for 8 cycles prior to surgery and an additional 10 cycles after surgery. Docetaxel will be co-administered during Cycles 1 to 4; chemotherapy during Cycles 5 to 8 will include 5-fluorouracil, cyclophosphamide, and epirubicin. Herceptin IV will be given on Day 1 of each 21-day cycle, as 8 milligrams per kilogram (mg/kg) for a loading dose during Cycle 1 and as 6 mg/kg during subsequent cycles.
Participants will receive 5-fluorouracil, 500 milligrams per meter-squared (mg/m^2) via IV bolus or infusion, on Day 1 of every 21-day cycle during Cycles 5 to 8.
Participants will receive cyclophosphamide, 500 mg/m^2 via IV bolus, on Day 1 of every 21-day cycle during Cycles 5 to 8.
Participants will receive docetaxel, 75 mg/m^2 via IV infusion on Day 1 of every 21-day cycle during Cycles 1 to 4.
Participants will receive epirubicin, 75 mg/m^2 via IV bolus or infusion, on Day 1 of every 21-day cycle during Cycles 5 to 8.
Herceptin will be administered as 8 mg/kg (loading dose during Cycle 1) and 6 mg/kg (subsequent cycles) via IV infusion on Day 1 of each 21-day cycle for a total of 18 cycles.
EXPERIMENTAL: Herceptin SC + Chemotherapy
Participants will receive Herceptin via SC injection for 8 cycles prior to surgery and an additional 10 cycles after surgery. Docetaxel will be co-administered during Cycles 1 to 4; chemotherapy during Cycles 5 to 8 will include 5-fluorouracil, cyclophosphamide, and epirubicin. Herceptin SC will be given on Day 1 of each 21-day cycle, as a 600-milligram (mg) fixed dose.
Participants will receive 5-fluorouracil, 500 milligrams per meter-squared (mg/m^2) via IV bolus or infusion, on Day 1 of every 21-day cycle during Cycles 5 to 8.
Participants will receive cyclophosphamide, 500 mg/m^2 via IV bolus, on Day 1 of every 21-day cycle during Cycles 5 to 8.
Participants will receive docetaxel, 75 mg/m^2 via IV infusion on Day 1 of every 21-day cycle during Cycles 1 to 4.
Participants will receive epirubicin, 75 mg/m^2 via IV bolus or infusion, on Day 1 of every 21-day cycle during Cycles 5 to 8.
Herceptin will be administered as fixed dose 600 mg SC on Day 1 of each 21-day cycle for a total of 18 cycles.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Observed Serum Trough Concentration (Ctrough) of Trastuzumab Prior to Surgery
Time Frame: Pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Pre-dose samples were obtained prior to surgery (Cycle 8). The observed Ctrough was recorded, averaged among all participants, and expressed in micrograms per milliliter (μg/mL).
Pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Percentage of Participants With Pathological Complete Response (pCR)
Time Frame: After surgery following eight cycles of Herceptin + chemotherapy (approximately 6 months from Baseline)
Participants were evaluated following eight cycles of treatment and after surgery to assess for pCR, defined as absence of neoplastic invasive cells in the breast according to pathologist examination. The percentage of participants with pCR was reported, and the 95% CI for one-sample binomial was constructed using the Pearson-Clopper method.
After surgery following eight cycles of Herceptin + chemotherapy (approximately 6 months from Baseline)

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Observed Ctrough of Trastuzumab After Surgery
Time Frame: Pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Pre-dose samples were obtained after surgery (Cycle 13). The observed Ctrough was recorded, averaged among all participants, and expressed in μg/mL.
Pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Predicted Ctrough of Trastuzumab Prior to Surgery
Time Frame: Pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Predicted Ctrough at pre-dose prior to surgery (Cycle 8) was determined on the basis of a population PK model from Study BP22023 (NCT00800436). The mean predicted Ctrough was expressed in μg/mL.
Pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Predicted Ctrough of Trastuzumab After Surgery
Time Frame: Pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Predicted Ctrough at pre-dose after surgery (Cycle 13) was determined on the basis of a population PK model from Study BP22023 (NCT00800436). The mean predicted Ctrough was expressed in μg/mL.
Pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Number of Participants With Ctrough of Trastuzumab >20 μg/mL Prior to Surgery
Time Frame: Pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Pre-dose samples were obtained prior to surgery (Cycle 8). The number of participants who had an observed Ctrough >20 μg/mL was reported.
Pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Number of Participants With Ctrough of Trastuzumab >20 μg/mL After Surgery
Time Frame: Pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Pre-dose samples were obtained after surgery (Cycle 13). The number of participants who had an observed Ctrough >20 μg/mL was reported.
Pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Maximum Serum Concentration (Cmax) of Trastuzumab Prior to Surgery
Time Frame: Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 7; on Days 2, 4, 8, 15 of Cycle 7; pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
PK samples were obtained prior to surgery (Cycle 7). The Cmax during Cycle 7 was recorded, averaged among all participants, and expressed in μg/mL.
Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 7; on Days 2, 4, 8, 15 of Cycle 7; pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Time of Maximum Serum Concentration (Tmax) of Trastuzumab Prior to Surgery
Time Frame: Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 7; on Days 2, 4, 8, 15 of Cycle 7; pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
PK samples were obtained prior to surgery (Cycle 7). The Tmax during Cycle 7 was recorded, averaged among all participants, and expressed in days.
Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 7; on Days 2, 4, 8, 15 of Cycle 7; pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Area Under the Concentration-Time Curve From 0 to 21 Days (AUC21d) of Trastuzumab Prior to Surgery
Time Frame: Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 7; on Days 2, 4, 8, 15 of Cycle 7; pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
PK samples were obtained prior to surgery (Cycle 7). Values were extrapolated beyond Day 15 to produce the area over the full 21-day cycle. The AUC21d value at Cycle 7 was calculated from trastuzumab concentration-time profiles using standard non-compartmental PK methods, averaged among all participants, and expressed in days multiplied by micrograms per milliliters (d*μg/mL).
Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 7; on Days 2, 4, 8, 15 of Cycle 7; pre-dose (0 hours) on Day 1 of Cycle 8 (cycle length of 21 days)
Cmax of Trastuzumab After Surgery
Time Frame: Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 12; on Days 2, 4, 8, 15 of Cycle 12; pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
PK samples were obtained after surgery (Cycle 12). The Cmax during Cycle 12 was recorded, averaged among all participants, and expressed in μg/mL.
Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 12; on Days 2, 4, 8, 15 of Cycle 12; pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Tmax of Trastuzumab After Surgery
Time Frame: Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 12; on Days 2, 4, 8, 15 of Cycle 12; pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
PK samples were obtained after surgery (Cycle 12). The Tmax during Cycle 12 was recorded, averaged among all participants, and expressed in days.
Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 12; on Days 2, 4, 8, 15 of Cycle 12; pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
AUC21d of Trastuzumab After Surgery
Time Frame: Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 12; on Days 2, 4, 8, 15 of Cycle 12; pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
PK samples were obtained after surgery (Cycle 12). Values were extrapolated beyond Day 15 to produce the area over the full 21-day cycle. The AUC21d value at Cycle 12 was calculated from trastuzumab concentration-time profiles using standard non-compartmental PK methods, averaged among all participants, and expressed in d*μg/mL.
Pre-dose (0 hours) and at end of 30-minute infusion (Herceptin IV only) on Day 1 of Cycle 12; on Days 2, 4, 8, 15 of Cycle 12; pre-dose (0 hours) on Day 1 of Cycle 13 (cycle length of 21 days)
Percentage of Participants With Total Pathological Complete Response (tpCR)
Time Frame: After surgery following eight cycles of Herceptin + chemotherapy (approximately 6 months from Baseline)
Participants were evaluated following eight cycles of treatment and after surgery to assess for tpCR, defined as absence of neoplastic invasive cells in the breast and axillary lymph nodes according to pathologist examination. The percentage of participants with tpCR was reported, and the 95% CI for one-sample binomial was constructed using the Pearson-Clopper method.
After surgery following eight cycles of Herceptin + chemotherapy (approximately 6 months from Baseline)
Percentage of Participants With Complete Response (CR) or Partial Response (PR) According to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.0, Among Those With Measurable Disease at Baseline
Time Frame: Tumor assessments at Baseline; on Day 1 of Cycles 3, 5, 7 (cycle length of 21 days); and at time of surgery following eight cycles of Herceptin + chemotherapy (approximately 6 months overall)
Tumor response was assessed using RECIST version 1.0. CR was defined as disappearance of all target lesions and short-axis reduction of any pathological lymph nodes to less than (<) 10 millimeters (mm) with no prior assessment of progressive disease (PD). PR was defined as greater than or equal to (≥) 30% decrease from Baseline in sum diameter (SD) of target lesions with no prior assessment of PD. PD was defined as ≥20% relative increase and ≥5 mm of absolute increase in the SD of target lesions, taking as reference the smallest SD recorded since treatment started; or appearance of 1 or more new lesions. The percentage of participants with overall response of CR or PR at the end of neoadjuvant treatment was reported, and the 95% CI for one-sample binomial was constructed using the Pearson-Clopper method.
Tumor assessments at Baseline; on Day 1 of Cycles 3, 5, 7 (cycle length of 21 days); and at time of surgery following eight cycles of Herceptin + chemotherapy (approximately 6 months overall)
Time to Response According to RECIST Version 1.0, Among Those With Measurable Disease at Baseline
Time Frame: Tumor assessments at Baseline; on Day 1 Cycles 3, 5, 7 (cycle length of 21 days); and at time of surgery following eight cycles of chemotherapy (approximately 6 months overall)
Tumor response was assessed using RECIST version 1.0. CR was defined as disappearance of all target lesions and short-axis reduction of any pathological lymph nodes to <10 mm with no prior assessment of PD. PR was defined as ≥30% decrease from Baseline in SD of target lesions with no prior assessment of PD. PD was defined as ≥20% relative increase and ≥5 mm of absolute increase in the SD of target lesions, taking as reference the smallest SD recorded since treatment started; or appearance of 1 or more new lesions. Time to response was defined as the time from first dose of study medication to the first assessment of CR or PR, which was the date the response was first documented by objective evidence, among participants with an overall response of CR or PR.
Tumor assessments at Baseline; on Day 1 Cycles 3, 5, 7 (cycle length of 21 days); and at time of surgery following eight cycles of chemotherapy (approximately 6 months overall)
Percentage of Participants Who Experienced a Protocol-Defined Event
Time Frame: Screening; Day 1 of Cycle 18 (cycle length of 21 days); and Months 6, 12, 24, 36, 48, 60 from last dose of Cycle 18; then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
Protocol-defined events included disease recurrence/progression (local, regional, distant, contralateral) or death from any cause. Imaging was performed at specified visits for up to 5 years after last dose. Thereafter, participants were followed for survival only. The percentage of participants who experienced a protocol-defined event at any time during the study was reported.
Screening; Day 1 of Cycle 18 (cycle length of 21 days); and Months 6, 12, 24, 36, 48, 60 from last dose of Cycle 18; then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
Event-Free Survival (EFS)
Time Frame: Screening; Day 1 of Cycle 18 (cycle length of 21 days); and Months 6, 12, 24, 36, 48, 60 from last dose of Cycle 18; then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
Protocol-defined events included disease recurrence or progression (local, regional, distant, contralateral) or death from any cause. Imaging was performed at specified visits for up to 5 years after last dose. Thereafter, participants were followed for survival only. EFS was estimated by Kaplan-Meier analysis and defined as the time from randomization to the first protocol-defined event.
Screening; Day 1 of Cycle 18 (cycle length of 21 days); and Months 6, 12, 24, 36, 48, 60 from last dose of Cycle 18; then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
Percentage of Participants Who Died
Time Frame: Continuously during treatment (up to 12 months); at Months 1, 3, 6 from last dose of Cycle 18 (cycle length of 21 days); then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
The percentage of participants who died at any time during the study was reported.
Continuously during treatment (up to 12 months); at Months 1, 3, 6 from last dose of Cycle 18 (cycle length of 21 days); then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
Overall Survival (OS)
Time Frame: Continuously during treatment (up to 12 months); at Months 1, 3, 6 from last dose of Cycle 18 (cycle length of 21 days); then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
OS was estimated by Kaplan-Meier analysis and defined as the time from randomization to death from any cause.
Continuously during treatment (up to 12 months); at Months 1, 3, 6 from last dose of Cycle 18 (cycle length of 21 days); then every 6 months until withdrawal for any reason (up to approximately 87 months overall)
Number of Participants With Anti-Drug Antibodies (ADAs) Against Trastuzumab
Time Frame: Baseline; pre-dose (0 hours) on Day 1 of Cycles 2, 5, 13, 18 (cycle length of 21 days); and Months 3, 6, 12, 18, 24, 30, 36, 42, 48, 54, 60 from last dose of Cycle 18
Participants provided PK samples for evaluation of anti-trastuzumab antibodies. The number of participants with "Treatment-induced ADAs" and "Treatment-enhanced ADA" against trastuzumab at any time during or after treatment was reported. Treatment-induced ADA = a participant with negative or missing Baseline ADA result(s) and at least one positive post-Baseline ADA result. Treatment-enhanced ADA = a participant with positive ADA result at Baseline who has one or more post Baseline titer results that are at least 0.60 titer unit greater than the Baseline titer result (four-fold increase of titer).
Baseline; pre-dose (0 hours) on Day 1 of Cycles 2, 5, 13, 18 (cycle length of 21 days); and Months 3, 6, 12, 18, 24, 30, 36, 42, 48, 54, 60 from last dose of Cycle 18
Number of Participants With ADAs Against Recombinant Human Hyaluronidase (rHuPH20)
Time Frame: Baseline; pre-dose (0 hours) on Day 1 of Cycles 2, 5, 13, 18 (cycle length of 21 days); and Months 3, 6, 12, 18, 24, 30, 36, 42, 48, 54, 60 from last dose of Cycle 18
Participants in the Herceptin SC arm provided PK samples for evaluation of anti-rHuPH20 antibodies. The number of participants with "Treatment-induced ADAs" and "Treatment-enhanced ADA" against rHuPH20 (an excipient unique to the SC formulation) at any time during or after treatment was reported. Treatment-induced ADA = a participant with negative or missing Baseline ADA result(s) and at least one positive post-Baseline ADA result. Treatment-enhanced ADA = a participant with positive ADA result at Baseline who has one or more post Baseline titer results that are at least 0.60 titer unit greater than the Baseline titer result (four-fold increase of titer).
Baseline; pre-dose (0 hours) on Day 1 of Cycles 2, 5, 13, 18 (cycle length of 21 days); and Months 3, 6, 12, 18, 24, 30, 36, 42, 48, 54, 60 from last dose of Cycle 18

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (ACTUAL)

October 16, 2009

Primary Completion (ACTUAL)

July 12, 2011

Study Completion (ACTUAL)

January 24, 2017

Study Registration Dates

First Submitted

July 30, 2009

First Submitted That Met QC Criteria

July 30, 2009

First Posted (ESTIMATE)

July 31, 2009

Study Record Updates

Last Update Posted (ACTUAL)

January 23, 2018

Last Update Submitted That Met QC Criteria

December 21, 2017

Last Verified

December 1, 2017

More Information

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Breast Cancer

Clinical Trials on 5-Fluorouracil

3
Subscribe