istitutofondazione ricerca piemonte per l’ cura del cancro ... · with her2-positive metastatic...

87
004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015 ________________________________________________________________________________________________ Confidential Page 1 of 87 Istituto per la Ricerca e la Cura del Cancro Fondazione del Piemonte per l’ Oncologia Open-Label, Phase II Study of Trastuzumab in Combination with Lapatinib or Pertuzumab in Combination with Trastuzumab in Patients with HER2-positive Metastatic Colorectal Cancer: the HERACLES Trial (HER2 Amplification for Colo-rectaL Cancer Enhanced Stratification) IMP Identifiers: Trastuzumab Lapatinb Pertuzumab Protocol Number: 004-IRCC-10IIS-12 EudraCT Number: 2012-002128-33 Protocol Version (Date): V3.0 - 18 June 2015 Sponsor: FPO_IRCC, Candiolo (Torino) CONFIDENTIAL This document contains confidential information belonging to Sponsor. Except as may be otherwise agreed to in writing, by accepting or reviewing these materials, you agree to hold such information in confidence and not to disclose it to others (except where required by applicable law), nor to use it for unauthorized purposes. In the event of actual or suspected breach of this obligation Sponsor should be promptly notified.

Upload: vuongnga

Post on 08-Nov-2018

219 views

Category:

Documents


0 download

TRANSCRIPT

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 1 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    Open-Label, Phase II Study of Trastuzumab in Combination with

    Lapatinib or Pertuzumab in Combination with Trastuzumab in Patients

    with HER2-positive Metastatic Colorectal Cancer: the HERACLES Trial

    (HER2 Amplification for Colo-rectaL Cancer Enhanced Stratification)

    IMP Identifiers: Trastuzumab

    Lapatinb

    Pertuzumab

    Protocol Number: 004-IRCC-10IIS-12

    EudraCT Number: 2012-002128-33

    Protocol Version (Date): V3.0 - 18 June 2015

    Sponsor: FPO_IRCC, Candiolo (Torino)

    CONFIDENTIAL

    This document contains confidential information belonging to Sponsor. Except as may be otherwise agreed

    to in writing, by accepting or reviewing these materials, you agree to hold such information in confidence

    and not to disclose it to others (except where required by applicable law), nor to use it for unauthorized

    purposes. In the event of actual or suspected breach of this obligation Sponsor should be promptly notified.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 2 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    SPONSOR SIGNATURE

    Paolo Maria Comoglio

    Sponsor Representative

    22/06/2015

    Signature Date

    STUDY CHAIR SIGNATURE

    Dr. Salvatore Siena

    Study Chair

    22/06/2015

    Signature Date

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 3 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    PRINCIPAL INVESTIGATOR AGREEMENT

    I have read the Protocol entitled Open-Label, Phase II Study of Trastuzumab in Combination with Lapatinib

    or Pertuzumab in Combination with Trastuzumab in Patients with HER2-positive Metastatic Colorectal

    Cancer: the HERACLES Trial (HER2 Amplification for Colo-rectaL Cancer Enhanced Stratification) and I

    agree to conduct the study as detailed herein and in compliance with ICH Guidelines for Good Clinical

    Practice and applicable regulatory requirements. I will provide all study personnel under my supervision with

    all information provided by the Sponsor and I will inform them about their responsibilities and obligations.

    Principal Investigator

    (printed name, Institution, Department and location)

    Signature Date

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 4 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    ADDITIONAL TRIAL PERSONNEL/SITE INFORMATION

    Sponsor Fondazione Piemonte Oncologia (FPO),

    Strada Provinciale 142 km 32, 10060 Candiolo (To)

    Phone: 011-993-3601

    Fax: 011-962-1525

    E-mail: [email protected]

    Sponsor authorized representative

    Prof. Paolo Maria Comoglio

    Strada Provinciale 142 km 32, 10060 Candiolo (To)

    Phone: 011-993-3601

    Fax: 011-962-1525

    E-mail: [email protected]

    Sponsor Medical officer

    Dr. Silvia Marsoni

    Strada Provinciale 142 km 32, 10060 Candiolo (To)

    Phone: 011-993-3926

    Fax: 011-962-1525

    E-mail: [email protected]

    Study Chair Dr. Salvatore Siena

    Divisione Oncologia Falck, Ospedale Niguarda Ca

    Granda, Piazza Ospedale Maggiore 3, 20162 Milano

    Phone: +39-02-6444.2291

    Fax: +39-02-6444.2957

    E-mail: [email protected]

    Principal Investigators Dr. Andrea Sartore Bianchi

    Divisione Oncologia Falck, Ospedale Niguarda Ca

    Granda, Piazza Ospedale Maggiore 3, 20162 Milano

    Phone: +39-02-6444.2291

    Fax: +39-02-6444.2957

    E-mail: [email protected]

    Dott. Francesco Leone

    Fondazione Piemonte Oncologia (FPO),

    Strada Provinciale 142 km 32, 10060 Candiolo (To)

    Phone: 011-993-3628

    Fax: 011-962-1525

    E-mail: [email protected]

    Dr. Zagonel Vittorina

    IOV, Istituto Oncologico Veneto, Padova

    Phone: +39 049 8215953 - 5909

    Fax: +39 049 8215890

    E-mail: [email protected]

    Dr. Carmine Pinto

    Unit Operativa di Oncologia Medica

    S. Orsola Malpighi (Bologna)

    Phone: 051-6362-204

    Fax: 051-6362-207

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 5 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    E-mail:[email protected]

    Dr. Fortunato Ciardiello

    Unit operativa complessa Oncologia Medica

    AOU Seconda Universit degli studi di Napoli .

    Phone: 081-5666-745

    Fax: 081-5666-732

    E-mail: [email protected]

    Contract Research Organization (CRO)

    CLIOSS Srl

    V.le Pasteur, 10

    20014 Nerviano (MI), ITALY

    Pharmacovigilance (SAE Reporting) Fondazione del Piemonte per lOncologia

    Dr. Cosimo Martino

    Strada Provinciale 142 km 32, 10060 Candiolo (To)

    Phone: 011-993-3842

    Fax: 011-993-3318

    E-mail: [email protected]

    Central laboratories for Pharmacogenomics

    (body fluids, tissue samples)

    Istituto di Anatomia Patologica Ospedale Niguarda Ca

    Granda,

    Dr. Emanuele Valtorta

    Piazza Ospedale Maggiore 3, 20162 Milano

    Phone: 02-6444-2702

    Fax:02-6444-2102

    E-mail: [email protected]

    Laboratorio di Genetica Molecolare Istituto per la Ricerca

    e la Cura del Cancro, Candiolo

    Prof. Alberto Bardelli

    S.P. 142 Km 3.95 Candiolo (TO)

    Phone: 011-993-3235

    Fax: 011-962-1525

    E-mail: [email protected]

    Laboratorio di Biochimica Clinica

    Piano Terra - Padiglione 9

    Dr. Laura Farioli, Dr. Simonetta Granata

    Ospedale Niguarda Ca Granda

    Piazza Ospedale Maggiore 3, 20162 Milano

    Phone + 39 02 6444 2428/2559

    Fax: + 39 02 6444 2901

    E-mail: [email protected] ;

    [email protected]

    mailto:[email protected]:[email protected]
  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 6 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    TABLE OF CONTENTS

    1 SYNOPSIS ............................................................................................................................................................. 9

    2 SCHEDULE OF EVENTS ..................................................................................................................................... 15

    3 ABBREVIATIONS AND DEFINITIONS OF TERMS ...................................................................................... 19

    4 BACKGROUND INFORMATION ...................................................................................................................... 21

    LAPATINIB ..................................................................................................................................................................... 21 TRASTUZUMAB .............................................................................................................................................................. 22 PERTUZUMAB ................................................................................................................................................................ 22

    5 STUDY RATIONALE ............................................................................................................................................ 23

    6 STUDY OBJECTIVES........................................................................................................................................... 23

    6.1 PRIMARY OBJECTIVE ......................................................................................................................................... 23 6.2 SECONDARY OBJECTIVE(S) ................................................................................................................................ 23 6.3 EXPLORATORY OBJECTIVE(S) ............................................................................................................................ 23

    7 STUDY ENDPOINTS ............................................................................................................................................ 24

    7.1 PRIMARY ENDPOINT .......................................................................................................................................... 24 7.2 SECONDARY ENDPOINT(S) ................................................................................................................................. 24 7.3 EXPLORATORY ENDPOINT(S) ............................................................................................................................. 24

    8 STUDY DESIGN .................................................................................................................................................... 24

    9 STUDY POPULATION ......................................................................................................................................... 24

    9.1 SUBJECT SELECTION .......................................................................................................................................... 24 9.1.1 Subject Inclusion Criteria ......................................................................................................................... 25 9.1.2 Subject Exclusion Criteria ........................................................................................................................ 25

    9.2 SCREENING FAILURES ........................................................................................................................................ 26 9.3 REPLACEMENTS ................................................................................................................................................. 26

    10 ENROLLMENT PROCEDURES ..................................................................................................................... 26

    11 STUDY TREATMENT ...................................................................................................................................... 27

    11.1 DESCRIPTION OF INVESTIGATIONAL PRODUCT (S) ............................................................................................. 27 11.1.1 Lapatinib ................................................................................................................................................... 27 11.1.2 Trastuzumab.............................................................................................................................................. 27 11.1.3 Pertuzumab ............................................................................................................................................... 27

    11.2 DRUG PREPARATION .......................................................................................................................................... 27 11.2.1 Instructions for trastuzumab reconstitution .............................................................................................. 27 11.2.2 Instructions for pertuzumab reconstitution ............................................................................................... 28

    11.3 TREATMENT DOSE AND SCHEDULE ................................................................................................................... 28 11.4 DURATION OF TREATMENT ................................................................................................................................ 30 11.5 DRUG ACCOUNTABILITY ................................................................................................................................... 30 11.6 SAFETY PROFILE OF INVESTIGATIONAL PRODUCTS ........................................................................................... 31

    11.6.1 Lapatinib Warnings and Precautions ....................................................................................................... 31 11.6.2 Trastuzumab Warnings and Precautions .................................................................................................. 32 11.6.3 Pertuzumab Warnings and Precautions.................................................................................................... 33

    11.7 TREATMENT INTERRUPTION AND DOSE MODIFICATIONS GUIDELINE IN CASE OF ADVERSE EVENT .................... 34 11.7.1 Dose Reductions ....................................................................................................................................... 34 11.7.2 Dose Delay and Dose reduction in case of Drug Related Toxicity ........................................................... 34

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 7 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    11.7.3 Infusion-Associated Symptoms and Allergic Reactions ................................................................................. 36 11.7.4 Incomplete Loading Dose of Trastuzumab and Pertuzumab .................................................................... 36 11.7.5 Continuation/discontinuation study treatment based on LVEF assessment ............................................. 37 11.7.6 Liver Chemistry Stopping Criteria ............................................................................................................ 38 11.7.7 Liver Chemistry Follow-up Criteria ........................................................................................................ 39 11.7.8 Management of Diarrhoea ........................................................................................................................ 40 11.7.9 Management of Dermatological events .................................................................................................... 40

    11.8 CONCOMITANT MEDICATIONS AND OTHER THERAPY ....................................................................................... 41

    12 SUBJECT WITHDRAWAL FROM STUDY PARTICIPATION ................................................................. 43

    13 TREATMENT ASSESSMENT ......................................................................................................................... 43

    13.1 SCREENING EVALUATIONS ................................................................................................................................ 43 13.2 ON STUDY EVALUATIONS.................................................................................................................................. 44 13.3 OFF TREATMENT EVALUATIONS ........................................................................................................................ 45 13.4 FOLLOW-UP EVALUATIONS ............................................................................................................................... 45 13.5 DETAILS OF INDIVIDUAL ASSESSMENTS ............................................................................................................ 45 13.6 PHARMACOGENOMICS STUDIES ......................................................................................................................... 46

    14 SAFETY ASSESSMENTS ................................................................................................................................. 47

    14.1 PRE-EXISTING CONDITION.................................................................................................................................. 47 14.2 ADVERSE EVENT ASSESSMENT .......................................................................................................................... 47 14.2 ADVERSE EVENT REPORTING PERIOD ............................................................................................................... 49 14.3 REPORTING PROCEDURES FOR ADVERSE EVENT ................................................................................................ 50 14.4 RECORDING ADVERSE EVENTS IN THE CASE REPORT FORMS ............................................................................ 51 14.5 CAUSALITY ASSESSMENT AND GRADING OF ADVERSE EVENT SEVERITY ......................................................... 51 14.6 PREGNANCY REPORTING ................................................................................................................................... 52 14.7 OVERDOSE ......................................................................................................................................................... 52 14.8 FOLLOW-UP OF UNRESOLVED ADVERSE EVENTS .............................................................................................. 53

    15 EFFICACY ASSESSMENTS ............................................................................................................................ 53

    15.1 DEFINITION OF EFFICACY PARAMETERS ............................................................................................................. 53

    16 STATISTICAL METHODS .............................................................................................................................. 53

    16.1 SAMPLE SIZE CALCULATION .............................................................................................................................. 53 16.2 STUDY POPULATION .......................................................................................................................................... 53 16.3 ANALYSIS .......................................................................................................................................................... 54

    16.3.1 Study Conduct and Subject Disposition ......................................................................................................... 54 16.3.2 Baseline Characteristics and treatment Group Comparability ..................................................................... 54 16.3.3 Treatment Analysis ........................................................................................................................................ 54 16.3.4 Safety Analysis ............................................................................................................................................... 54 16.3.5 Pharmacogenomics Analysis (or any other ancillary analysis if applicable) ................................................ 55 16.3.6 Efficacy Analysis ............................................................................................................................................ 55

    17 QUALITY CONTROL AND QUALITY ASSURANCE ................................................................................ 55

    17.1 MONITORING ..................................................................................................................................................... 55 17.2 AUDITING .......................................................................................................................................................... 56 17.3 LABORATORY REQUIREMENTS .......................................................................................................................... 56

    18 DATA HANDLING AND RECORD KEEPING ............................................................................................. 56

    18.1 CASE REPORT FORM (CRF) ........................................................................................................................... 56 18.2 DATA HANDLING ............................................................................................................................................... 56 18.3 RECORD RETENTION .......................................................................................................................................... 57

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 8 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    19 ETHICAL CONSIDERATION ......................................................................................................................... 57

    19.1 INSTITUTIONAL REVIEW BOARD(IRB)/ INDEPENDENT ETHICS COMMITTEE (IEC) AND COMPETENT AUTHORITY (CA) 57 19.2 ETHICAL CONDUCT OF THE TRIAL ...................................................................................................................... 57 19.3 INFORMED CONSENT ......................................................................................................................................... 57

    20. STUDY DISCONTINUATION CRITERIA .................................................................................................... 58

    21 LIABILITY AND INSURANCE ....................................................................................................................... 58

    22 CONFIDENTIALITY OF INFORMATION AND PUBLICATION OF RESULTS ................................... 59

    23 REFERENCES ................................................................................................................................................... 59

    24 APPENDIX 59

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 9 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    1 SYNOPSIS

    Protocol Title: Open-Label, Phase II Study of Trastuzumab in Combination with Lapatinib or

    Pertuzumab in Combination with Trastuzumab in Patients with HER2-positive

    Metastatic Colorectal Cancer: the HERACLES Trial (HER2 Amplification for

    Colo-rectaL Cancer Enhanced Stratification)

    Protocol Number:

    IMPS: Lapatinib

    Oral formulation

    250 mg tablets

    Glaxo Smith Kline

    Trastuzumab

    i.v. formulation

    440 or 150 mg/vial

    Roche

    Pertuzumab

    i.v. formulation

    30 mg/mL

    Roche

    Participating countries Italy

    List of study centres Ospedale Niguarda Ca Granda, Milano; FPO, Fondazione Piemonte Oncologia,

    Candiolo, Torino, IOV, Istituto Oncologico Veneto, Padova; Policlinico S. Orsola

    Malpighi, Bologna, Universit Federico II, Napoli.

    Background Information and

    Study Rationale

    Metastatic colorectal carcinoma (mCRC) is the second or third most common

    cancer in developed countries and remains a nearly universally fatal disease;

    despite the treatment advances made in the last decade, the 5-year survival for

    patients with mCRC remains indeed only 10%1. The development monoclonal

    antibodies (moAbs) directed against the epidermal growth factor receptor (EGFR)

    has provided additional therapeutic options for patients with mCRC progressing

    through chemotherapy, and may increase the efficacy of chemotherapy when

    given in combination2. Response rates in the range of 13%-17% are achievable by

    EGFR-targeted moAbs in tumours without mutations in codon 12 or 13 of the

    KRAS gene, whereas only 0%-1.2% of the KRAS mutant tumours respond to this

    therapy3. Nevertheless, even in KRAS wild-type CRCs, about 40% of the

    previously untreated and about 60%-70% of the previously treated do not respond

    to anti-EGFR treatment4. Additional detection of NRAS, BRAF,and PIK3CA exon

    20 mutations and loss of PTEN protein may further enhance selection of patients

    but elucidation of other mechanism(s) by which tumor cell resistance is acquired

    in this setting and identification of druggable molecular targets to overcome

    resistance are clearly a priority of clinical research, since there are no other

    available therapeutic options for patients progressing during or after standard

    medical therapies.

    In a recent study by our group, we exploited direct transfer xenografts of mCRC

    surgical specimens in mice (xenopatients) to discover novel determinants of

    therapeutic response and new druggable driver oncoproteins6. This approach,

    combining the flexibility of preclinical analysis with the informative value of

    population-based studies, led to the identification of HER2 amplification as an

    additional molecular marker of resistance to EGFR-targeted therapy with

    cetuximab in xenopatients harbouring mCRC tumors for which other known

    molecular alterations conferring resistance were excluded, i.e., KRAS, NRAS,

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 10 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    BRAF, and PIK3CA mutations. In this defined subset, HER2 amplification was

    detected in 4 of 11 cases (36%), suggesting a greater frequency of HER2

    amplification in cetuximab-resistant cases and its progressive enrichment along

    with refinement of genetic selection. Indeed, data from literature show that HER2

    amplification occurs in a small percentage (2%-3%) of genetically unselected

    colorectal cancers7, 8, 9

    , but its incidence increases in KRAS wild-type colorectal

    cancer patients that displayed de novo resistance to anti-EGFR therapies (13.6%)

    and possibly rises to a summit in KRAS/NRAS/BRAF/PIK3CA WT tumors

    resistant to EGFR-targeted therapies6.

    A proof-of-concept, multi-arm study in HER2-amplified xenopatients revealed

    that combinations of the dual small molecule EGFR/HER2 inhibitor lapatinib and

    the anti-HER2 moAb trastuzumab or lapatinib and the HER-dimerization inhibitor

    moAb pertuzumab induced overt, long-lasting tumor regressions, while

    monotherapy with lapatinib led to disease stabilization and either monotherapy

    trastuzumab or pertuzumab were ineffective6. The combination of pertuzumab and

    trastuzumab has also a strong rationale for treatment of HER2-amplified tumors,

    since recent clinical observations have demonstrated that combining these two

    moAbs together yields synergistic results in HER2 positive breast cancers that

    progressed during prior trastuzumab therapy19

    , suggesting a cooperative

    mechanism of inhibition. Currently, the latter combination is also being explored

    in our xenopatients platform (Trusolino, personal communication).

    Based on these findings, the combinations of trastuzumab and lapatinib or

    pertuzumab and trastuzumab appear to have a sounded rationale for the treatment

    of those mCRC harbouring amplification of the HER2 oncogene. This hypothesis-

    driven trial will test whether these two distinct anti-HER2 therapy combinations

    would produce a response rate 30% in chemorefractory mCRC patients, for

    whom further effective treatment remains an unmet clinical need. Safe doses for

    both trastuzumab combinations have been already established in the clinic for

    gastric and breast cancer in different clinical settings. The combination of

    pertuzumab and trastuzumab has been tested in breast cancer patients (pertuzumab

    840 mg/loading dose, followed by 420 mg q3wks, plus trastuzumab 4 mg/kg

    loading dose, then 2 mg/kg weekly or 8 mg/kg loading dose, then 6 mg/kg

    q3wks19

    .

    Primary Objective Define the antitumor activity of the anti-HER2 combinations of lapatinib +

    trastuzumab and pertuzumab + trastuzumab given to two separate, sequential

    cohorts of patients with chemo-refractory advanced disease and HER2 amplified

    tumours.

    Secondary Objective(s) 1. Define the safety profile of the two combinations

    2. Define the Progression Free Survival (PFS)

    Exploratory Objectives 1. To determine whether selected tumor biomarkers evaluated in tumor specimens,

    including but not limited to mutations of effectors downstream of HER-family

    receptors (such as KRAS, BRAF and PIK3CA) or related tyrosine kinase

    receptors, have association with response/resistance to experimental treatment

    2. To determine whether selected tumor biomarkers as evaluated from circulating

    tumor DNA, including but not limited to mutations of effectors downstream of

    HER-family receptors (KRAS, BRAF, and PIK3CA) or related tyrosine kinase

    receptors, evolve during tumor progression

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 11 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    Primary Endpoint Objective Response Rate according to RECIST 1.1 criteria

    Secondary Endpoint(s) 1. Description of the frequency and severity of Adverse Events based on the NCI CTCAE V4.0

    2. PFS

    Exploratory Endpoints 1. Correlation between selected tumor biomarkers evaluated in tumor specimens (including but not limited to mutations of effectors downstream of HER-family

    receptors - such as KRAS, BRAF, and PIK3CA, or related tyrosine kinase

    receptors), and tumor response/resistance to experimental treatment

    2. Comparison of tumor biomarkers status in blood samples collected before start

    of treatment, with that collected during treatment and after progression

    Study Design This is a Phase II multi-center 2-sequential cohorts trial, designed to assess the

    objective response rate of the anti HER2 monoclonal antibody trastuzumab, used

    in combination with either the small molecule tyrosine kinase inhibitor lapatinib

    (Cohort A) or the monoclonal antibody pertuzumab (Cohort B), in advanced

    disease CRC patients harbouring an amplified HER2 tumor (SISH).

    Treatment Cohort A:

    - lapatinib 1000 mg daily per os

    - trastuzumab 4 mg/kg iv load, followed by 2 mg/kg iv weekly

    Cohort B:

    - pertuzumab 840 mg/kg iv load, followed by 420 mg/kg iv Q3weeks

    - trastuzumab 8 mg/kg iv load, followed by 6 mg/kg on day 1 of each

    subsequent 3 week cycle

    Treatment Duration Patients enrolled in both cohorts will receive study medication until disease

    progression, unacceptable toxicity, withdrawal of consent or death, whichever

    come first.

    Supportive Therapy N.A.

    Efficacy Assessments Objective tumor response in target and non-target lesions will be assessed

    according to RECIST 1.1 criteria.

    Tumor response will be assessed starting on week 8 and every 8 weeks thereafter.

    Tumor response MUST be confirmed 4 + 1week from first assessment of

    response.

    Safety Assessments Frequency and severity of Adverse Events will be recorded according to the NCI

    CTCAE V4.0.

    Pharmacogenomics

    Assessments

    Tumor specimens (primary tumor and/or metastases) must be available in a

    mandatory manner for all patients for evaluation of HER2 status (as per inclusion

    criteria) and evaluation of selected tumor biomarkers including but not limited to

    mutations of effectors downstream of HER-family receptors (such as KRAS,

    BRAF and PIK3CA) or related tyrosine kinase receptors, with the aim of

    evaluating their potential association with response/resistance to experimental

    treatment.

    All patients will undergo a blood test for retrieving circulating tumor DNA

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 12 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    (ctDNA) at selected time-points before and during treatment for determining

    whether the status of selected tumor biomarkers, including but not limited to

    molecular alterations of effectors downstream of HER-family receptors or related

    tyrosine kinase receptors (such as mutations of KRAS, BRAF and PIK3CA)

    evolve during tumor progression by comparing different ctDNA samples. Blood

    samples will be also analysed for determination of circulating serum levels of the

    extra-cellular domain of HER2 (HER2 ECD) as a non invasive surrogate measure

    of changes in tumor HER2 during treatment.

    Sample Size For each cohort the sample size has been calculated basing on the Fleming & Hern

    single stage design, and under the following identical assumptions:

    1. Standard proportion of eligible patients which would respond to a

    chemotherapy (H0) = 10%

    2. Minimum required proportion of patients expected to respond to the

    combination (H1) = 30%

    Given =0.05 and power=0.85, 27 patient (N) are required in each trial cohort

    (Part A 27 + Part B 27 = 54 patients overall).

    Each combination (A or B) will be considered positive if at least 6 responses/27

    patients are observed.

    In March 2015, HERACLES met the primary endpoint, since we observed 8

    responses in the first 23 evaluable patients (1CR+7PR).

    In May 2015 we have enrolled the 27th patient required to complete cohort A, but

    in the screening procedures we found other 6 potentially eligible HER2 amplified

    patients. We plan to extend cohort A enrollment from 27 to 33 patient for ethical

    reason, in order to not deprive these patients of a potential effective and very well

    tolerated treatment.

    Under the same statistical assumptions, extending the cohort A sample size from

    27 to 33 will require 7/33 instead of 6/27 OR in order to declare the study

    positive; as of June 2015 we already have 8 OR the study will be positive in any

    case.

    Inclusion Criteria

    (valid for Cohorts A & B)

    Subjects must meet all the following inclusion criteria to be eligible for enrolment

    into the study:

    1. Histological/confirmed adenocarcinoma of the colon or rectum with

    metastatic disease not amenable to salvage surgery

    2. Pathology mandatory requirements:

    a. the original tumour specimen must be KRAS WT and SISH/FISH positive or IHC 3+ positive in more than 50% cells.

    Note: for immunohistochemistry a positive staining (3+) is defined as an

    intense membrane staining which can be circumpherential, basolateral,

    or lateral of the tumor cells.

    b. the original paraffin block or a minimum of 15 polarized unstained

    slides from the original paraffin block must be made available to the

    Pathology Core within 15 days from registration.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 13 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    3. Age 18.

    4. ECOG PS 0-1.

    5. Measurable disease as defined by RECIST 1.1 criteria.

    6. Progression (PD) while on treatment, or within 6 months from therapy with

    approved standard drugs.

    7. Unless otherwise contraindicated patients should have received and failed

    the following previous therapies for mCRC: fluoropirimidines, oxaliplatin,

    irinotecan, cetuximab or panitumumab containing regimens. Bevacizumab is

    allowed.

    8. Adequate haematological function as defined by: ANC 1.5 x 109/L, platelet

    count 100 x 109/L, haemoglobin 10 g/dL.

    9. Adequate renal function, as defined by: creatinine 1.5 x UNL.

    10. Adequate hepatobiliary function, as defined by the following baseline liver

    function tests:

    o total serum bilirubin 1.5 upper normal limit (UNL)

    o alanine aminotransferase (ALT), aspartate aminotransferase (AST)

    2.5xUNL

    o alkaline phosphatase (AP) 2.5xUNL; if total alkaline phosphatase (AP)

    > 2.5xUNL, alkaline phosphatase liver fraction must be 2.5xUNL

    11. Adequate contraception for all fertile patients.

    12. Negative pregnancy test.

    Exclusion criteria

    (valid for Cohorts A & B)

    Subjects meeting any of the following criteria must not be enrolled in the study:

    1. Radiotherapy 4 weeks prior to enrolment

    2. Other chemotherapy or biological therapy treatment 4 weeks prior to

    enrolment

    3. Symptomatic brain metastases

    4. Active infection

    5. Gastro-intestinal abnormalities, inability to take oral medication, any

    condition affecting absorption

    6. Impaired cardiac function including any of the following: uncontrolled

    hypertension (systolic >150 mmHg and/or diastolic > 100 mmHg) or

    clinically significant (ie active) cardiovascular disease: cerebrovascular

    accident/stroke or myocardial infarction within 6 months prior to first study

    medication; unstable angina; chronic heart failure (CHF) of New York Heart

    Association (NYHA) Grade II or higher; or serious cardiac arrhythmia

    requiring medication, baseline Left Ventricular Ejection Fraction (LVEF)

    55% measured by echocardiography (ECHO)

    7. Major surgery in the two weeks prior to entering the clinical trial

    8. Concurrent treatment with any other anti-cancer therapy

    9. History of another neoplastic disease (except basal cell carcinoma of the skin

    or uterine cervix carcinoma in situ adequately treated), unless in remission

    for 5 years

    10. Patient unable to comply with the study protocol owing to psychological,

    social or geographical reasons

    11. Pregnant and lactating women

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 14 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    12. Patients with history of hypersensitivity to either IMPs or excipients

    13. Men and women of childbearing potential who are not using an effective

    method of contraception

    14. Participation in another clinical trial or treatment with any investigational

    product within 4 weeks prior to inclusion in this study.

    15. Subjects who have current active hepatic or biliary disease (with exception

    of patients with Gilbert's syndrome, asymptomatic gallstones, liver

    metastases or stable chronic liver disease per investigator assessment)

    16. Patient with unresolved hypokalaemia, hypomagnesemia or hypocalcaemia

    Patients replacement Patients not evaluable for efficacy (Section 16.2) will be replaced

    Planned study timelines FPI Cohort A: August 2012

    LPI Cohort A (27 pts): May 2015

    Expected FPI Cohort A (6pts expansion): July 2015

    Expected LPI Cohort A (6pts expansion): September 2015

    Expected LPO Cohort A: August 2016

    Expected FPI Cohort B: September 2015

    Expected LPI Cohort B: December 2016

    Expected LPO Cohort B: December 2017

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 15 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    2 SCHEDULE OF EVENTS

    COHORT A

    Assessment/Event

    Pre

    study 1

    Cycle Q3wks

    Treatment day

    End of

    Treatment

    Visit 2

    F-up

    visit3

    D -14 to 0 D1

    (D22) D8 D15

    4 wks

    from last

    treatment

    Q8 wks

    Signed informed consent x

    Demographic data x

    Baseline condition x

    Medical History/Diagnosis /Prior TX x

    Physical examination 4 x x x x x

    Vital signs x x x x x

    ECOG PS x x x x x

    Hematology 5 x x x

    Chemistry 6 x x x

    Tumor markers7 x x

    Pregnancy test x

    ECG8 x Only if clinically indicated

    LVEF9 x Q3 months x

    Plasma Sample for Companion study 10 x x Q2 weeks x x

    Serum Sample for HER2 ECD determination14 x Q8 weeks x x

    Tumor assessment11 x Q8 weeks x x

    Concomitant medication Throughout the study

    Adverse Events Monitoring Throughout the study

    Lapatinib administration12 Daily continously

    Trastuzumab administration 13 x x x

    See numbered Footnotes for further details on Schedule of Events.

    1. Within 2 weeks of starting therapy, except X-rays and scans (4 weeks). Laboratory assessments performed within 72 hrs of study treatment will be considered good also for D1.

    2. End of Treatment visit to be performed approximately 4 weeks after last treatment cycle

    3. Follow-up visit for patients without documented PD at the time of treatment withdrawal: to be performed every 8 weeks until PD or another anti-cancer therapy is initiated, whichever comes first. Evaluations to be performed

    according to clinical practise

    4. Physical examination including body skin exam according to Appendix 5

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 16 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    5. Hematology: hemoglobin, hematocrit, platelet count, total white blood cell count (WBC) and differential (neutrophil count, lymphocyte, monocyte, eosinophil, and basophil counts), red blood cell count (RBC). To be

    performed within 72 hours from the day reported on the schedule of assessment

    6. Chemistry: sodium, potassium, chloride, bicarbonate, creatinine, calcium, albumin, total bilirubin, total protein, glucose, alkaline phosphatase, AST, ALT urea or BUN. To be performed within 72 hours from the day reported

    on the schedule of assessment

    7. Tumor Markers: CEA, CA19.9. to be performed on Day 1 of each 3 weeks cycle

    8. 12 lead electrocardiogram (ECG)

    9. After the basal assessment echocardiograpic monitoring of the potential cardio toxicity will be done every three months or if clinically indicated according to Section 11.7.5.1 and 11.7.5.2. At EOT visit echocardiograpic

    monitoring has to repeated if previous echocardiogram was performed > 3 months before

    10. Companion study Cohort A as specified in section 13.6. Samples to be taken at different time points according to the following indication: patients will be sampled at baseline, every two weeks (i.e wks 2, wks 4, wks 6, etc),

    and at disease progression

    11. A CT scan or MRI should be performed within 4 weeks prior to the first dose and every 8 weeks during treatment until PD; however in order to fulfil RECIST, the FIRST occurrence of response MUST be reconfirmed

    after 4 + 1 week later. Patients without documented PD at the time of treatment withdrawal should continue to

    have disease assessments done every 8 weeks until PD or another anti-cancer therapy is initiated, whichever

    comes first

    12. Lapatinib administration for patients enrolled in Cohort A: 1000 mg (4 tablets)

    13. Trastuzumab administration for patients enrolled in Cohort A: trastuzumab 2 mg/kg iv weekly. A 4 mg/kg trastuzumab loading dose is required on day 1.

    14. Companion study Cohort A as specified in section 13.6. Samples to be taken at different time points according to the following indication: patients will be sampled at baseline, every eight weeks (i.e wks 8, wks 16, wks 24,

    etc), and at disease progression.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 17 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    COHORT B

    Assessment/Event

    Pre

    study 1

    Cycle Q3wks

    Treatment day

    End of

    Treatment

    Visit 2

    F-up

    visit3

    D -14 to 0 D1

    (D22) D8 D15

    4 wks

    from last

    treatment

    Q8 wks

    Signed informed consent x

    Demographic data x

    Baseline condition x

    Medical History/Diagnosis /Prior TX x

    Physical examination4 x x x

    Vital signs x x x

    ECOG PS x x x

    Hematology 5 x x x

    Chemistry 6 x x x

    Tumor markers7 x x

    Pregnancy test x

    ECG8 x Only if clinically indicated

    LVEF9 x Q3 months x

    Plasma Sample for Companion study Cohort B10 x x x x

    Serum Sample for HER2 ECD determination14 x Q8 weeks x x

    Tumor assessment11 x Q8 weeks x x

    Concomitant medication Throughout the study

    Adverse Events Monitoring Throughout the study

    Trastuzumab administration Cohort B 12 x

    Pertuzumab administration13 x

    See numbered Footnotes for further details on Schedule of Events.

    1. Within 2 weeks of starting therapy, except X-rays and scans (4 weeks). Laboratory assessments performed within 72 hrs of study treatment will be considered good also for D1.

    2. End of Treatment visit to be performed approximately 4 weeks after last treatment cycle

    3. Follow-up visit for patients without documented PD at the time of treatment withdrawal: to be performed every 8 weeks until PD or another anti-cancer therapy is initiated, whichever comes first. Evaluations to be performed

    according to clinical practise

    4. Physical examination including body skin exam according to Appendix 5

    5. Hematology: hemoglobin, hematocrit, platelet count, total white blood cell count (WBC) and differential (neutrophil count, lymphocyte, monocyte, eosinophil, and basophil counts), red blood cell count (RBC). To be

    performed within 72 hours from the day reported on the schedule of assessment

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 18 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    6. Chemistry: sodium, potassium, chloride, bicarbonate, creatinine, calcium, albumin, total bilirubin, total protein, glucose, alkaline phosphatase, AST, ALT urea or BUN. To be performed within 72 hours from the day reported

    on the schedule of assessment

    7. Tumor Markers: CEA, CA19.9. to be performed on Day 1 of each 3 weeks cycle

    8. 12 lead electrocardiogram (ECG)

    9. After the basal assessment echocardiograpic monitoring of the potential cardio toxicity will be done every three months or if clinically indicated according to Section 11.7.5.1 and 11.7.5.2. At EOT visit echocardiograpic

    monitoring has to repeated if previous echocardiogram was performed > 3 months before

    10. Companion study Cohort B as specified in section 13.6. Samples to be taken at different time points according to the following indication: Patients will be sampled at baseline, on Day 1 of each 3 weeks cycle, and at disease

    progression

    11. A CT scan or MRI should be performed within 4 weeks prior to the first dose and every 8 weeks during treatment until PD; however in order to fulfil RECIST, the FIRST occurrence of response MUST be reconfirmed after 4 + 1

    week later. Patients without documented PD at the time of treatment withdrawal should continue to have disease

    assessments done every 8 weeks until PD or another anti-cancer therapy is initiated, whichever comes first

    12. Trastuzumab administration for patients enrolled in Cohort B: trastuzumab 8 mg/kg iv loading dose on day 1 of the first treatment cycle, followed by 6 mg/kg iv of each subsequent 3 weeks cycle

    13. Pertuzumab administration for patients enrolled in Cohort B: pertuzumab 840 mg/kg iv loading dose on day 1of the first treatment cycle, followed by 420 mg/kg iv on day 1 of each subsequent 3 week cycle.

    14. Companion study Cohort B as specified in section 13.6. Samples to be taken at different time points according to the following indication: patients will be sampled at baseline, every eight weeks (i.e wks 8, wks 16, wks 24, etc),

    and at disease progression.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 19 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    3 ABBREVIATIONS AND DEFINITIONS OF TERMS

    AE Adverse Event

    ALT Alanine Aminotransferase

    ANC Absolute Neutrophil Count

    AP Alkaline Phopsphatase

    ARDS Acute Respiratory Distress Syndrome

    AST Aspartate Aminotransferase

    BSA Body Surface Area

    BUN Blood Urea Nitrogen

    CA Competent Authority

    CHF Congestive Heart Failure

    CR Complete Response

    CTM Clinical Trial Monitor

    CRC Colorectal Carcinoma

    CRF (eCRF) Case Report Form (Electronic Case Report Form

    CT Computerized Tomography

    CTCAE Common Terminology Criteria For Adverse Events

    CYP3A4 Human Cytochrome P450 34A

    EC Ethics Committee

    ECG Electrocardiography, Electrocardiogram

    ECHO Echocardiogram

    ECOG Eastern Cooperative Oncology Group

    EE Efficacy Evaluable

    FPPE Formalin Fixed Paraffin Embedded

    GCP Good Clinical Practices

    ICF Informed Consent Form

    ICH International Conference On Harmonisation

    IEC Independent Ethics Committe

    IMP Investigational Medical Product

    IRB Institutional Review Board

    i.v. Intravenous

    LDH Lactate Dehydrogenase

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 20 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    LVEF Left Ventricular Ejection Fraction

    MedDRA Medical Dictionary For Regulatory Activities

    MRI Magnetic Resonance Imaging

    NOAEL No observed adverse effect level

    NYHA New York Heart Association

    PD Progressive disease

    PFS Progression Free Survival

    PG Pharmacogenomics

    PI Principal investigator

    PLTs Platelets / thrombocytes

    p.o. Per os / oral

    PR Partial response

    PS Performance status

    PT Preferred Term

    QA Quality assurance

    RBC Red Blood Cell

    RD Recommended Dose

    RECIST Response evaluation criteria in solid tumors

    SAE Serious adverse event

    SD Stable disease

    SE Safety evaluable

    SISH Silver Immunohistochemistry

    SOC System Organ Class

    SOP Standard Operating Procedure

    UNL Upper Normal Limit

    WBC White Blood Count / White Blood Cells

    WHO World Health Organization

    wks weeks

    WT Wild Type

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 21 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    4 BACKGROUND INFORMATION

    Metastatic colorectal carcinoma (mCRC) is the second or third most common cancer in both men and

    women in developed countries and remains a nearly universally fatal disease; despite the treatment advances

    made in the last decade, the 5-year survival for patients with mCRC remains indeed only 10%1. First line

    chemotherapy for this disease is typically a 5-FU/oxaliplatin/folate containing regimen bevacizumab, with

    irinotecan-containing regimens such as FOLFIRI used second line, but long-standing responses following

    either of these regimens are rare, and the survival of patients failing both these regimens is usually measured

    in months. The development monoclonal antibodies directed against the epidermal growth factor receptor

    (EGFR), such as panitumumab and cetuximab, has provided additional therapeutic options for patients

    progressing through chemotherapy and may increase the efficacy of chemotherapy when given in

    combination2. Although initial response rates of about 10% were seen with these agents in patients with

    chemorefractory mCRC, it was subsequently discovered that higher response rates in the range of 13%-17%

    were achievable in tumours without mutations in codon 12 or 13 of the KRAS gene, whereas only 0%-1.2%

    of the KRAS mutant tumours responded to therapy3. Nevertheless, even in KRAS wild-type CRCs, about

    40% of the previously untreated and about 60%-70% of the previously treated do not respond to anti-EGFR

    treatmentand additional detection of NRAS, BRAF and PIK3CA exon 20 mutations and loss of PTEN

    protein4 or better discrimination among KRAS mutations by excluding G13D carriers

    5 may further enhance

    selection of patients, but elucidation of other mechanism(s) by which tumor cell resistance is acquired in this

    setting and identification of druggable molecular targets to overcome resistance are an unmet need in mCRC,

    since there are no other available therapeutic options for patients progressing through chemotherapy.

    In a recent article by our group, we exploited direct transfer xenografts of mCRC surgical specimens in mice

    (xenopatients) to discover novel determinants of therapeutic response and new druggable driver

    oncoproteins6. This approach, combining the flexibility of preclinical analysis with the informative value of

    population-based studies, led to the identification of HER2 amplification as an additional molecular marker

    of resistance to EGFR-targeted therapy with cetuximab in xenopatients harbouring tumors for which other

    known molecular alterations conferring resistance were excluded (i.e. KRAS, NRAS, BRAF and PIK3CA

    mutations). In this defined subset, HER2 amplification was detected in 4 of 11 cases (36%), suggesting a

    greater frequency of HER2 amplification in cetuximab-resistant cases and its progressive enrichment along

    with refinement of genetic selection. Indeed, data from literature show that HER2 amplification occurs in a

    small percentage (2%3%) of genetically unselected colorectal cancers7, 8, 9

    , but its incidence increases in

    KRAS wild-type colorectal cancer patients that displayed de novo resistance to anti-EGFR therapies (13.6%)

    and possibly rises to a summit in KRAS/NRAS/BRAF/PIK3CA WT tumors resistant to EGFR-targeted

    therapies6.

    Lapatinib

    Lapatinib is a reversible TKI that potently inhibits both ErbBl and ErbB2 tyrosine kinase activity. There are

    several theoretical advantages of an inhilbitor of both ErbBl and ErbB2 compared to inhibitors of either one

    of these tyrosine kinases alone. While small molecule Inhibitors of ErbBl will inhibit ErbBl homodimers,

    they may not be as effective at inhibiting heterodimers containing ErbB I and ErbB2. For example, treatment

    with gefitinib, a TKI of ErbBl, plus the antibody trastuzumab induces a greater apoptotic effect than either

    inhibitor alone in ErbB2-overexpressing human breast cancer cell lines SKBR-3 and BT-17410

    . Additionally,

    lapatinib exhibits greater growth inhibition of TGF -activated colon cancer cells than antagonists targeting

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 22 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    only ErbB1 or ErbB2 receptors11

    . These data suggest that a dual ErbB1 and ErbB2 inhibitor may provide

    improved therapeutic benefit as compared with Inhibitors that target either receptor alone12

    .

    A phase II study, EGF20008, of single agent lapatinib In subjects with advanced or metastatic breast cancer

    who had progressed while receiving anthracyclines, taxanes, capecitabine and trastuzumab-containing

    regimens demonstrated an Investigator determined response rate of 4.3% in 140 subjects whose tumors

    overexpress the ErbB2 protein by immunohistochemistry (IHC) or demonstrate ErbB2 gene amplification by

    fluorescence in situ hybridization (FISH). Furthermore, the investigator determined clinical benefit rate

    (defined as partial response, complete response or stable disease > 6 months) was noted at 5.7%. These data

    indicate that in a heavily pre-treated population where there exists an unmet medical need, single agent

    lapatinib elicits a response.

    Trastuzumab

    Trastuzumab is a humanized monoclonal antibody directed at the HER2 receptor and is indicated for the

    treatment of patients with HER2-positive breast cancer both in the adjuvant and metastatic setting. The

    addition of trastuzurnab to standard chemotherapy increases time to progressive disease or the length of

    progression free survival (PFS), and improves survival when given with chemotherapy to women with

    HER2-positive breast cancer13, 14

    .

    Clinical benefits are greatest in patients with HER2 gene amplification and or tumors strongly

    overexpressing HER2, graded 3+ by immunohistochemistry (IHC: see Herceptin Package Insert,

    November 2006). Trastuzumab is well tolerated both as a single agent and in combination with standard

    chemotherapy 15, 14

    . The most significant adverse event (AE) observed in patients who received trastuzumab

    was cardiac dysfunction, reflected by asymptomatic decreases in left ventricular ejection fraction (LVEF)

    and, less frequently, by clinically symptomatic congestive heart failure (CHF). Risk factors for cardiac

    failure in the setting of trastuzumab treatment include co-administration with anthracycline-based

    chemotherapy, increasing age, declining LVEF during treatment to below the lower limit of normal, and the

    use of anti-hypertensive medications16

    .

    Pertuzumab

    Pertuzumab is a fully humanized monoclonal antibody based on the human immunoglobulin (I)Gl (k)

    framework sequences and consisting of two heavy chains (449 residues) and two light chains (214 residues).

    Similar to trastuzumab, pertuzumab is directed against the extracellular domain of HER2: however, it differs

    from trastuzumab in the epitope-binding regions of the light chain (12 ammo acid differences) and heavy

    chain (29 amino acid differences). As a result, pertuzumab binds to an epitope within what is known as sub-

    domain 2 of HER2, while the epitope for trastuzumab is localized to sub-domain 417,18

    .

    Pertuzumab acts by blocking the dimerization of HER2 with other HER family members, including HER1

    {epidermal growth factor receptor [EGFR]), HER3, and HER4. As a result, pertuzumab inhibits ligand-

    initiated intra cellular signaling through two major signal pathways. MAP-kinase and PI3-kinase. Inhibition

    of these signaling pathways can result in growth arrest and apoptosis, respectively19

    . Data from a clinical trial

    of lapatinib support the hypothesis that HER2 plays an active role in tumor biology, with progression of

    MBC occurring even after treatment with trastuzumab20

    . The results obtained suggest that a broader

    blockade of HER2 through interruption of heterodimerization may provide clinical benefit.

    Due to the different binding sites of pertuzumab and trastuzumab. ligand-activated downstream signaling is

    blocked by pertuzumab, but not by trastuzumab. Due to their complementary modes of action, there is a

    potential role for the combination of pertuzumab and trastuzumab m HER2-overexpressing diseases.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 23 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    5 STUDY RATIONALE

    A proof-of-concept, multi-arm study in HER2-amplified xenopatients revealed that combinations of the dual

    small molecule EGFR/HER2 inhibitor lapatinib and HER-dimerization inhibitor moAb pertuzumab induced

    overt, long-lasting tumor regressions, while monotherapy with lapatinib led to disease stabilization6 and

    either monotherapy with trastuzumab or pertuzumab was ineffective (Bertotti et al.6 and Trusolino, personal

    communication). The combination of pertuzumab and trastuzumab has also a strong rationale for treatment

    of HER2-amplified tumors, since recent clinical observations have demonstrated that combining these two

    moAbs together yields synergistic results in HER2 positive breast cancers that progressed during prior

    trastuzumab therapy19

    , suggesting a cooperative mechanism of inhibition. Currently, this combination is also

    being explored in our xenopatients platform (Trusolino, personal communication).

    Based on these findings, the combinations of trastuzumab and lapatinib or pertuzumab and trastuzumab

    appear to have a sounded rationale for the treatment of those mCRC harbouring amplification of the HER2

    oncogene. This hypothesis-driven trial will test whether these two distinct anti-HER2 therapy combinations

    would produce a response rate 30% in chemorefractory mCRC patients, for whom further effective

    treatment remains an unmet clinical need. Safe doses for both trastuzumab combinations have been already

    established in the clinic: association of trastuzumab and lapatinib has been evaluated in breast cancer

    (trastuzumab 4 mg/kg loading dose followed by 2 mg/kg weekly and lapatinib 1000 mg qd)21

    and is being

    currently evaluated in other clinical settings. Combination of pertuzumab and trastuzumab has also been

    tested in breast cancer patients (pertuzumab 840 mg/loading dose, followed by 420 mg q3wks, plus

    trastuzumab 4 mg/kg loading dose, then 2 mg/kg weekly or 8 mg/kg loading dose, then 6 mg/kg q3wks)19

    .

    6 STUDY OBJECTIVES

    6.1 Primary Objective

    Define the antitumor activity of the anti-HER2 combinations of lapatinib + trastuzumab and pertuzumab +

    trastuzumab given to two separate, sequential cohorts of patients with chemo-refractory advanced disease

    and HER2 amplified tumours.

    6.2 Secondary Objective(s)

    - Define the safety profile of the two combinations

    - Define the PFS of the combinations lapatinib + trastuzumab and pertuzumab + trastuzumab given to two separate, sequential cohorts of patients with chemo-refractory advanced disease and HER2 amplified

    tumours.

    6.3 Exploratory Objective(s)

    To determine whether selected tumor biomarkers evaluated in tumor specimens, including but not limited to molecular alterations of effectors downstream of HER-family receptors (such as mutations of

    KRAS, BRAF and PIK3CA) or related tyrosine kinase receptors, have association with

    response/resistance to experimental treatment

    To determine whether selected tumor biomarkers as evaluated from circulating tumor DNA, including but not limited to molecular alterations of effectors downstream of HER-family receptors (such as

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 24 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    mutations of KRAS, BRAF and PIK3CA) or related tyrosine kinase receptors, evolve during tumor

    progression

    7 STUDY ENDPOINTS

    7.1 Primary Endpoint

    Objective response rate according to RECIST criteria

    7.2 Secondary Endpoint(s)

    Description of the frequency and severity of Adverse Events based on the NCI CTCAE V4.0

    PFS

    7.3 Exploratory Endpoint(s)

    Correlation between selected tumor biomarkers (including but not limited to molecular alterations of effectors downstream of HER-family receptors - such as mutations of KRAS, BRAF and PIK3CA, or

    related tyrosine kinase receptors), and tumor response/resistance to experimental treatment

    Comparison of tumor biomarkers status in blood samples collected before start of treatment, with that collected during treatment and after progression

    8 STUDY DESIGN

    This is a phase II, open label, two-sequential cohorts, multicenter study.

    Patients with advanced colon rectal cancer (CRC) harbouring an amplified HER2 tumor (SISH), progressed

    while on treatment or within 6 months from last therapy, are eligible to participate in the study and will be

    treated with the anti HER2 monoclonal antibody trastuzumab, used in combination with either the small

    molecule tyrosine kinase inhibitor lapatinib (Cohort A) or the monoclonal antibody pertuzumab (Cohort B).

    Approximately 27 patients are foreseen to be enrolled sequentially in each cohort.

    Patients will receive study medication until disease progression, unacceptable toxicity, withdrawal of consent

    or death.

    Main objective of the study is the evaluation of the response rate, followed by the progression free survival

    and the safety profile of the 2 combinations. As exploratory objectives the study foresees to evaluate the

    correlation between selected biomarkers from tumor specimens and tumor response, and to evaluate whether

    those tumor biomarkers (circulating tumor DNA) evolve during tumor progression.

    Patient enrolled in cohort A will receive lapatinib 1000 mg daily per os and trastuzumab 4 mg/kg iv loading

    dose on D1, followed by 2 mg/kg iv weekly. Patients enrolled in Cohort B will receive pertuzumab 840

    mg/kg iv loading dose on the first day of cycle 1, followed by 420 mg/kg iv on D1 of each subsequent 3

    weekly cycle and trastuzumab 8 mg/kg iv loading dose, followed by 6 mg/kg iv on Day 1 of each subsequent

    3 weeks cycle.

    9 STUDY POPULATION

    9.1 Subject Selection

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 25 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    9.1.1 Subject Inclusion Criteria

    Subjects must meet all the following inclusion criteria to be eligible for enrolment into the study:

    1. Histological/confirmed adenocarcinoma of the colon or rectum with metastatic disease not amenable to salvage surgery

    2. Pathology mandatory requirements:

    a. the original tumour specimen must be KRAS WT and SISH/FISH positive or IHC 3+ positive in more than 50% cells.

    Note: for immunohistochemistry a positive staining (3+) is defined as an intense membrane

    staining which can be circumpherential, basolateral, or lateral of the tumor cells.

    b. the original paraffin block or a minimum of 15 polarized unstained slides from the original paraffin block must be made available to the Pathology Core within 15 days from registration.

    3. Age 18

    4. ECOG PS 0-1

    5. Measurable disease as defined by RECIST 1.1 criteria.

    6. Progression (PD) while on, or within 6 months from therapy with approved standard drugs

    7. Unless otherwise contraindicated patients should have received and failed the following previous therapies for mCRC: fluoropirimidines, oxaliplatin, irinotecan, cetuximab or panitumumab

    containing regimens. Bevacizumab is allowed

    8. Adequate haematological function as defined by: ANC 1.5 x 109/L, platelet count 100 x 109/L,

    haemoglobin 10 g/dL

    9. Adequate renal function, as defined by: creatinine 1.5 x UNL

    10. Adequate hepatobiliary function, as defined by the following baseline liver function tests:

    o total serum bilirubin 1.5 upper normal limit (UNL)

    o alanine aminotransferase (ALT), aspartate aminotransferase (AST) 2.5xUNL

    o alkaline phosphatase (AP) 2.5xUNL; if total alkaline phosphatase (AP) > 2.5xUNL, alkaline

    phosphatase liver fraction must be 2.5xUNL

    11. Adequate contraception for all fertile patients

    12. Negative pregnancy test

    9.1.2 Subject Exclusion Criteria

    Subjects meeting any of the following criteria must not be enrolled in the study:

    1. Radiotherapy 4 weeks prior to enrolment

    2. Other chemotherapy or biological therapy treatment 4 weeks prior to enrolment

    3. Symptomatic brain metastases

    4. Active infection

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 26 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    5. Gastro-intestinal abnormalities, inability to take oral medication, any condition affecting absorption

    6. Impaired cardiac function including any of the following: uncontrolled hypertension (systolic >150 mmHg and/or diastolic > 100 mmHg) or clinically significant (ie active) cardiovascular disease:

    cerebrovascular accident/stroke or myocardial infarction within 6 months prior to first study

    medication; unstable angina; chronic heart failure (CHF) of New York Heart Association (NYHA)

    Grade II or higher (See Appendix 4); or serious cardiac arrhythmia requiring medication, baseline

    Left Ventricular Ejection Fraction (LVEF) 55% measured by echocardiography (ECHO)

    7. Major surgery in the two weeks prior to entering the clinical trial

    8. Concurrent treatment with any other anti-cancer therapy

    9. History of another neoplastic disease (except basal cell carcinoma of the skin or uterine cervix carcinoma in situ adequately treated), unless in remission for 5 years

    10. Patient unable to comply with the study protocol owing to psychological, social or geographical reasons

    11. Pregnant and lactating women

    12. Patients with history of hypersensitivity to either IP or excipients

    13. Men and women of childbearing potential who are not using an effective method of contraception

    14. Participation in another clinical trial or treatment with any investigational product within 4 weeks prior to inclusion in this study.

    15. Subjects who have current active hepatic or biliary disease (with exception of patients with Gilbert's syndrome, asymptomatic gallstones, liver metastases or stable chronic liver disease per investigator

    assessment)

    16. Patient with unresolved hypokalaemia, hypomagnesemia or hypocalcaemia

    9.2 Screening failures

    For all screened patients, a Subject Screening Log will be completed and in case of screening failure the

    reason(s) for failure will be documented. CRFs have to be completed only for registered patients.

    9.3 Replacements

    Patients not evaluable for efficacy according to the definition described in section 16.2 will be replaced.

    10 ENROLLMENT PROCEDURES

    Before any screening procedure all patients must sign a written informed consent for the study. The

    following logs must be maintained at each study site and kept in the Investigator File: a Subject Screening

    and Enrolment Log, to document the identification of subjects who are screened and enrolled; a Subject

    Identification Code List for all subjects registered to maintain the correlation with the patient's full

    identification data (name, surname - confidential). Before registration in the study, all the pre-treatment

    evaluations must be reported on the "Registration Form and faxed to FPO fax number +39 011-993.3318 or

    e-mailed to [email protected]. Upon review of all inclusion/exclusion criteria, if the patient is eligible,

    mailto:[email protected]
  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 27 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    a progressive Registration Number, is centrally assigned by FPO and provided to the Investigator by fax or

    by E-mail on a Confirmation of Registration Form.

    Any controversial eligibility assessment will be discussed with the Study Chair.

    11 STUDY TREATMENT

    11.1 Description of Investigational Product (s)

    11.1.1 Lapatinib

    Lapatinib Ditosylate Monohydrate Tablets, 250 mg, are oval, biconvex, orange, filmcoated tablets that are

    debossed on one side with FG HLS. The tablets contain 410 mg of lapatinib ditosylate monohydrate,

    equivalent to 250 mg lapatinib free base per tablet. Lapatinib may also be supplied as oval, biconvex, tan

    tablets, with no markings. Refer to the Investigators Brochure (IB) for information regarding the physical

    and chemical properties of the drug substance and list of excipients. Lapatininb is provided free of charge by

    Glaxo Smith Kline.

    11.1.2 Trastuzumab

    Trastuzumab (Herceptin) is supplied in 150 mg single-dose vials or 440 mg multidose vials containing white

    to pale yellow lyophilized powder for concentrate for infusion. Reconstituted Trastuzumab concentrate

    contains trastuzumab 21 mg/mL. The concentrate for infusion will be accompanied by a solvent vial (for use

    with 440mg vial) and bacteriostatic water for injection containing 1.1% benzyl alcohol. Trastumuzumab has

    to be stored at 2C8C. Refer to the Product Information Sheet for information regarding the physical and

    chemical properties of trastuzumab as well as drug substance and list of excipients. Trastuzumab is provided

    free of charge by Roche.NTIAUM2005/00071/04

    1040

    11.1.3 Pertuzumab

    Pertuzumab is provided as a single-use formulation containing 30 mg/mL pertuzumab formulated in 20 mM

    L-histidine (pH 6.0), 120 mM sucrose, and 0.02% polysorbate 20. Each 20-cc vial contains approximately

    420 mg of pertuzumab (14.0 mL/vial). For further details, see the pertuzumab Investigator Brochure (IB).

    Pertuzumab is provided free of charge by Roche. UM2005/00071/04

    11.2 Drug preparation

    11.2.1 Instructions for trastuzumab reconstitution

    1) Using a sterile syringe, slowly inject 7.2 ml of sterile water for injections in the vial containing the

    lyophilised trastuzumab, directing the stream into the lyophilised cake.

    2) Swirl vial gently to aid reconstitution. DO NOT SHAKE!

    Slight foaming of the product upon reconstitution is not unusual. Allow the vial to stand undisturbed for

    approximately 5 minutes. The reconstituted trastuzumab results in a colourless to pale yellow transparent

    solution and should be essentially free of visible particulates.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 28 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    Determine the volume of the solution required:

    Cohort A: based on a loading dose of 4 mg trastuzumab/kg body weight, or a subsequent weekly dose of 2

    mg trastuzumab/kg body weight:

    Volume (ml) =Body weight (kg) x dose (4 mg/kg for loading or 2mg/kg for maintenance)

    21 (mg/ml, concentration of reconstituted solution)

    Cohort B: based on a loading dose of 8 mg trastuzumab/kg body weight, or a subsequent 3-weekly dose of

    6 mg trastuzumab/kg body weight:

    Volume (ml) = Body weight (kg) x dose (8 mg/kg for loading or 6mg/kg for maintenance)

    21 (mg/ml concentration of reconstituted solution)

    The appropriate amount of solution should be withdrawn from the vial and added to an infusion bag

    containing 250 ml of 0.9 % sodium chloride solution. Do not use with glucose-containing solutions. The bag

    should be gently inverted to max the solution in order to avoid foaming. Parenteral solutions should be

    inspected visually for particulates and discoloration prior to administration. Once the infusion is prepared it

    should be administered immediately. If diluted aseptically, it may be stored for 24 hours (do not store above

    30C). No incompatibilities between trastuzumab and polyvinylchloride or polyethylene bags have been

    observed.

    11.2.2 Instructions for pertuzumab reconstitution

    Because the pertuzumab formulation does not contain a preservative, the vial seal may only be punctured

    once. Any remaining solution should be discarded.

    The indicated volume of pertuzumab solution should be withdrawn from the vials and added to a 250-cc IV

    bag of 0.9% sodium chloride injection. Gently invert the bag to mix the solution. DO NOT SHAKE

    VIGOROUSLY. Visually inspect the solution for particulates and discoloration prior to administration. The

    entire volume within the bag should be administered as a continuous IV infusion. The volume contained in

    the administration tubing should be completely flushed using a 0.9% sodium chloride injection.

    The solution of pertuzumab for infusion diluted in PVC or non-PVC polyolefin bags containing 0.9%

    sodium chloride injection may be stored at 2C8C for up to 24 hours prior to use. Diluted pertuzumab has

    been shown to be stable for up to 24 hours at room temperature. However, since diluted pertuzumab contains

    no preservative, the aseptically diluted solution should be stored refrigerated (2C8C) for no more than 24

    hours.

    11.3 Treatment Dose and Schedule

    Table 1 below provides study therapies with their respective dosage and the mode of administration:

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 29 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    Table 1. Dose and Mode of administration

    Drug Dosage Time administration

    Cohort A

    Lapatinib 1000 mg (4 Tablets) Daily

    Approximately the same time of day, preferably either at

    least 1 hour before or 1 hour after breakfast

    Trastuzumab

    Loading Dose

    4 mg/Kg iv infusion

    On Day 1 of first week.

    Approximately 90 minutes infusion

    Subsequent therapy 2 mg/kg iv infusion Weekly.

    Approximately 30 minutes infusion

    Cohort B

    Pertuzumab

    Loading Dose

    840 mg/Kg iv infusion

    On Day 1 of the first cycle.

    Approximately 60 ( 10) minutes infusion

    Subsequent therapy 420 mg/kg iv infusion On Day 1 of each 3 week cycle

    Approximately 30 minutes infusion

    Trastuzumab

    Loading Dose

    8 mg/Kg iv infusion

    To administer after pertuzumab, on Day 1 of the 1st cycle

    Approximately 90 minutes infusion

    Subsequent therapy 6 mg/kg iv infusion To administer after pertuzumab, on Day 1 of each

    subsequent 3 week cycle.

    Approximately 30 ( 10) minutes infusion

    Notes on administration of Lapatinib

    Lapatinib should not be taken with grapefruit or grapefruit juice. Ingestion of grapefruit or grapefruit juice is

    not permitted during the study

    If subject vomits after taking lapatinib therapy, the subject should be instructed not to retake the dose Subject

    should take the next schedule dose. If vomiting persists , then the subject should contact the investigator.

    Notes on administration of pertuzumab

    1. Initial infusions of pertuzumab will be administered over 60 ( 10) minutes and patients observed for a further 60 minutes from the end of infusion for infusion-associated symptoms such as fever, chills etc.

    Interruption or slowing of the infusion may reduce such symptoms. If the infusion is well tolerated,

    subsequent infusions may be administered over 30 ( 10) minutes, with patients observed for a further

    30 minutes.

    2. Pertuzumab administration may be delayed to assess or treat AEs such as cardiac AEs, myelosuppression, or other events. No dose reduction will be allowed for pertuzumab.

    3. To avoid or reduce the risk of infusion reactions a premedication must be administered before each pertuzumab infusion according to dosage and timing reported in the following table:

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 30 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    Chlorphenamine Maleate Dexamethasone Ranitidine

    Dose 10 mg 4 mg 50 mg

    Route im iv iv

    Timing 30 minutes before infusion After Chlorphenamine 15 minutes before infusion

    Notes on administration of trastuzumab

    1. Trastuzumab administration should be follow by an observational period in line with approved local Product Information.

    2. Trastuzumab administration may be delayed to assess or treat AEs such as cardiac AEs, myelosuppression, or other events. No dose reduction will be allowed for trastuzumab.

    3. To avoid or reduce the risk of infusion reactions a premedication must be administered before each trastuzumab infusion according to dosage and timing reported in the following table:

    Chlorphenamine Maleate Dexamethasone Ranitidine

    Dose 10 mg 4 mg 50 mg

    Route im iv iv

    Timing 30 minutes before infusion After Chlorphenamine 15 minutes before infusion

    11.4 Duration of Treatment

    Duration of treatment will depend on the response to therapy. Patients enrolled in both cohorts will receive

    study medication until disease progression, unacceptable toxicity, withdrawal of consent or death, whichever

    come first.

    11.5 Drug Accountability

    The Investigator and/or a pharmacist or other appropriate individual designated by the Investigator, should

    maintain records of the amount of investigational drug delivered to the trial site, the inventory at the site, the

    amount of drug administered to each subject and the unused drug to be destroyed locally. The pharmacist, or a delegated person at the site, will be responsible for handling study drug(s) preparation

    of the appropriate doses to be administered, and completion of drug accountability forms.

    Study drug(s) must be handled and administered strictly in accordance with the protocol and/or the specific

    manual and will be stored in a limited access area or in a locked cabinet under appropriate storage

    conditions. Study drug(s) should be administered under the supervision of the investigator or co-investigator,

    only to subjects participating in the study in accordance with the approved protocol.

    Unused study drug(s) must be available for verification by the sponsors site monitor during on site

    monitoring visits.

  • 004-IRCC-10IIS-12 Protocol Version 3.0 - 18 June 2015

    ________________________________________________________________________________________________

    Confidential Page 31 of 87

    Istituto per la Ricerca e la Cura del Cancro

    Fondazione del Piemonte per l Oncologia

    11.6 Safety Profile of Investigational Products

    11.6.1 Lapatinib Warnings and Precautions

    Cardiac dysfunction

    Lapatinib has been associated with decreases in LVEF. Caution should be taken if lapatinib is to be

    administered to patients with conditions that could impair left ventricular function. LVEF should be

    evaluated in all patients prior to initiation of treatment with lapatinib to ensure that the patient has a baseline

    LVEF that is within the institutions normal limits and > 55%. LVEF should continue to be evaluated during

    treatment with lapatinib to ensure that LVEF does not decline to an unacceptable level (see Section 11.7.5

    Continuation/discontinuation study treatment based on LVEF assessment)

    A small, concentration dependent increase in QTc interval was observed in an uncontrolled, open-label dose

    escalation study of lapatinib in advanced cancer patients. Caution should be taken if lapatinib is administered

    to patients who have or may develop prolongation of QTc. These conditions include patients with

    hypokalemia or hypomagnesemia, congenital long QT syndrome, patients taking anti-arrhythmic medicines

    or other medicinal products that lead to QT prolongation. Hypokalemia, hypocalcaemia or hypomagnesemia

    should be corrected prior to lapatinib administration.

    Pulmonary dysfunction

    Lapatinib has been associated with reports of Interstitial Lung Disease (ILD) and pneumonitis. Patients

    should be monitored for pulmonary symptoms indicative of ILD/pneumonitis.

    Hepatic impairment

    Lapatinib is metabolised in the liver. Moderate and severe hepatic impairment have been associated,

    respectively, with 56% and 85% increases in systemic exposure Hepatotoxicity (ALT or AST >3 times the

    UNL and total bilirubin >1.5 times the UNL) has bee