Protocol No: ECCT/22/04/02 Date of Protocol: 29-04-2021

Study Title:

A PHASE Ia/Ib DOSE-ESCALATION AND DOSE- EXPANSION STUDY EVALUATING THE SAFETY, PHARMACOKINETICS, AND ACTIVITYOF GDC-6036 AS A SINGLE AGENT AND IN COMBINATION WITH OTHER ANTI-CANCER THERAPIES IN PATIENTS WITH ADVANCED OR METASTATIC SOLID TUMORS WITH A KRAS 

 

Study Objectives:

This Phase Ia/Ib study will evaluate the safety, pharmacokinetics, immunogenicity (as applicable for study biotherapeutics), preliminary activity, and biomarkers of GDC-6036 as a single agent (Arm A) and in combination with other anti-cancer therapies in patients with advanced or metastatic solid tumors with a KRAS G12C mutation. Combination therapies will include atezolizumab (Arm B), cetuximab (Arm C), bevacizumab (Arm D), erlotinib (Arm E), and GDC-1971 (Arm F) in NSCLC, CRC, and other solid tumors.  

SAFETY OBJECTIVE (PRIMARY STUDY OBJECTIVE)

The safety objective for this study is to evaluate the safety of GDC-6036 as a single agent and in combination with other anti-cancer therapies on the basis of the following endpoints:

·Incidence and severity of adverse events, with severity determined according to National Cancer Institute Common Terminology Criteria for Adverse Events, Version 5.0 (NCI CTCAE v5.0)

· Incidence and nature of DLTs

· Change from baseline in targeted vital signs

· Change from baseline in targeted clinical laboratory test results

· Change from baseline in targeted ECG parameters

PHARMACOKINETIC OBJECTIVES

The pharmacokinetic (PK) objectives for this study are to characterize the PK profile of

GDC-6036 when administered as a single agent and in combination with other

anti-cancer therapies and to characterize the PK profile of these anti-cancer therapies when administered in combination with GDC-6036, on the basis of the following endpoints:

·     Plasma concentrations of GDC-6036, erlotinib, and GDC-1971 at specified timepoints

·     Serum concentrations of atezolizumab, cetuximab, and bevacizumab at specified timepoints

The exploratory PK objectives for this study are as follows:

·     To evaluate potential relationships between drug exposure and the safety and activity of GDC-6036 as a single agent and in combination with other anti-cancer therapies

·     To evaluate the exposure of potential circulating metabolites of GDC-6036 following a single or repeat oral dose(s) of GDC-6036 as a single agent or in combination with other anti-cancer therapies

·     To assess ex vivo plasma protein binding of GDC-6036 and its impact on pharmacokinetics (Arm A only)

·     To evaluate the effect of GDC-6036 on plasma levels of 4b-hydroxy cholesterol, an endogenous biomarker of CYP3A4 induction (Arm A only)

·     To evaluate the effect of a standard meal taken within 30 minutes of a GDC-6036 oral dose on the pharmacokinetics of GDC-6036 (Arm A only)

·     To assess potential relationships between drug exposures and the safety and activity of GDC-1971 in combination with GDC-6036

·   To evaluate the exposure of potential circulating metabolites of GDC-1971 following a single or repeat oral dose(s) of GDC-1971 in combination with GDC-6036

 IMMUNOGENICITY OBJECTIVE

The exploratory immunogenicity objective for this study is to evaluate the immune response to study biotherapeutics on the basis of the following endpoints:

·     Prevalence of anti-drug antibodies (ADAs) at baseline and incidence of ADAs after initiation of study treatment

·      Evaluation of safety, efficacy, PD, and PK endpoints by ADA status

ACTIVITY OBJECTIVES

The activity objective for this study is to make a preliminary assessment of the activity of GDC-6036 as a single agent and in combination with other anti-cancer therapies on the basis of the following endpoints:

·    Objective response rate (ORR), defined as the proportion of patients with a complete response (CR) or PR on two consecutive occasions ³ 4 weeks apart, as determined by the investigator according to Response Evaluation Criteria in Solid Tumors, Version 1.1 (RECIST v1.1)

·    Duration of response (DOR), defined as the time from the first occurrence of a documented objective response to disease progression or death from any cause during the study (whichever occurs first), as determined by the investigator according to RECIST v1.1

·    Progression-free survival (PFS), defined as the time from first treatment at Cycle 1

Day 1 to the first occurrence of disease progression or death from any cause during the study (whichever occurs first), as determined by the investigator according to RECIST v1.1

BIOMARKER OBJECTIVE

The exploratory biomarker objective for this study is to identify and/or evaluate biomarkers that are potentially predictive of response to GDC-6036 as a single agent or in combination with other anti-cancer therapies (i.e., predictive biomarkers), early surrogates of activity, associated with progression to a more severe disease state

(i.e., prognostic biomarkers), associated with intrinsic or acquired resistance to KRAS

G12C inhibitors (e.g., GDC-6036), associated with susceptibility to developing adverse events or can lead to improved adverse event monitoring or investigation (i.e., safety biomarkers), can provide evidence of GDC-6036 activity as a single agent or in combination with other anti-cancer therapies (i.e., pharmacodynamic [PD] biomarkers), or can increase the knowledge and understanding of disease biology and drug safety. Corresponding biomarker endpoints include the following:

·      Relationship between exploratory biomarkers in blood, plasma, and tumor tissue (listed in Section 4.5.6) and safety, PK, activity, or other biomarker endpoints

ADDITIONAL OBJECTIVE

An additional objective for this study is to identify a recommended Phase II dose and regimen for GDC-6036 when administered as a single agent and when administered in combination with other anti-cancer therapies on the basis of any of the following endpoints:

·     Relationship between GDC-6036 exposure (PK parameters) and safety and activity endpoints

·     Relationship between tumor pharmacodynamic effects of GDC-6036 and safety and activity endpoints

Laymans Summary:

This is a first-in-human Phase Ia/Ib, open-label, multicenter dose-escalation and dose-expansion study designed to evaluate the safety, pharmacokinetics, and preliminary activity of GDC-6036 as a single agent and in combination with other anti-cancer therapies in patients with advanced or metastatic solid tumors that harbor the KRAS G12C mutation. The combination therapies in this study are atezolizumab (Arm B), cetuximab (Arm C), bevacizumab (Arm D), erlotinib (Arm E), and GDC-1971 (Arm F) in NSCLC, CRC, and solid tumors. The study is designed with the intention to include new, additional treatment arms during study conduct (via protocol amendments) to explore combinations of GDC-6036 with other anti-cancer therapies based on emerging nonclinical and clinical data with GDC-6036, other KRAS G12C inhibitors, or evolving standard-of-care treatment. Anticipated future combinations with GDC-6036 may include RTK/RAS/MAPK pathway-targeting therapies, agents that target compensatory pathways that may mediate intrinsic or acquired resistance to treatment, immunotherapies, agents that modulate the tumor microenvironment, and standard-of-care agents to further explore safety, pharmacokinetics, and preliminary activity.

1 his is a first--in--human Phase Ia/Ib, open--label, multicenter dose--escalation and dose-expansion study designed to evaluate the safety, pharmacokinetics, and preliminary activity of GDC--6036 as a single agent and in combination with other anti--cancer therapies in patients with advanced or metastatic solid tumors that harbor the KRAS G12C mutation. The combination therapies in this study are atezolizumab (Arm B), cetuximab (Arm C), bevacizumab (Arm D), erlotinib (Arm E), and GDC--1971 (Arm F), and inavolisib (Arm G) in NSCLC, CRC, and solid tumors. The study is designed with the intention to include new, additional treatment arms during study conduct (via protocol amendments) to explore combinations of GDC--6036 with other anti--cancer therapies based on emerging nonclinical and clinical datawith GDC--6036, and other KRAS G12C inhibitors, (e.g., mechanisms of response and resistance to GDC-6036 or other KRAS G12C inhibitors), or evolving standard--of--care treatment. Anticipated future combinations with GDC-6036 may include RTK/RAS/MAPK pathway--targeting therapies, agents that target compensatory pathways that may mediate intrinsic or acquired resistance to treatment, immunotherapies, agents that modulate the tumor microenvironment, and standard--of--care agents to further explore safety, pharmacokinetics, and preliminary activity.
Abstract of Study:

The Kirsten rat sarcoma viral oncogene homolog (KRAS) is a central component of the RAS/MAPK signal transduction pathway, an intracellular network of proteins that transmit extracellular growth factor signals to regulate cell proliferation, differentiation, and survival. Mutations in KRAS can result in alterations at several amino acids, including glycine 12 (G12), glycine 13, and glutamine 61, commonly found in solid tumors and associated with tumorigenesis and aggressive tumor growth (Der et al. 1982; Parada et al. 1982; Santos et al. 1982; Taparowsky et al. 1982; Capon et al. 1983). Oncogenic KRAS mutations that result in the change from G12 to cysteine (G12C) are prevalent in non-small cell lung cancer (NSCLC) (~12%), colorectal cancer (CRC) (~4%), and other tumor types (£ 4%) (Bailey et al. 2016; Campbell et al. 2016; Giannakis et al. 2016; Hartmaier et al. 2017; Jordan et al. 2017).

Advanced stage tumors harboring the KRAS G12C mutation (hereafter referred to as KRAS G12C-positive tumors), including NSCLC, CRC, and other solid tumors, are incurable and carry a poor prognosis (Roman et al. 2018; Wan et al. 2019). In addition, patients with advanced stage KRAS G12C-positive cancers may derive limited benefit from select chemotherapies and targeted therapies, thus, restricting effective available treatment options (Roman et al. 2018).

Initial systemic treatment options for advanced stage or metastatic NSCLC (without known oncogenic drivers that have available targeted therapies) include PD-1/PD-L1 inhibitors with or without chemotherapy (Gong et al. 2018).  Subsequent treatment options may include platinum-containing chemotherapy combinations followed by single-agent chemotherapy with limited duration of disease control (NCCN 2020a).

Although a minority of patients achieve long-term disease control, in general, advanced stage or metastatic NSCLC remains an incurable disease.  Recent data suggest that KRAS mutation status may be associated with response to single-agent PD-1 inhibitor therapy and that chemotherapy plus a PD-1 inhibitor may be effective regardless of KRAS mutation status (Gadgeel et al. 2019; Herbst et al. 2019).

For advanced or metastatic CRC, systemic treatment can consist of combinations of active agents or select individual agents and includes 5-fluorouracil/leucovorin, capecitabine, irinotecan, oxaliplatin, bevacizumab, cetuximab, panitumumab,ziv-aflibercept, ramucirumab, regorafenib, trifluridine-tipiracil, pembrolizumab, nivolumab, ipilimumab, and encorafenib. Treatment is selected based on the goals of care, type and timing of prior therapy, mutational profile of the tumor, anticipated toxicity profile, and patient’s performance status (NCCN 2020b). Specifically, KRAS mutations were associated with resistance to anti-EGFR therapies (Lièvre et al. 2006; Karapetis et al.2008; Van Cutsem et al. 2009; Misale et al. 2012). Thus, patients whose cancers harbor KRAS mutations are not eligible for treatment with cetuximab or panitumumab (NCCN 2020b) and treatment options remain limited.

Other tumor types with the KRAS G12C mutation include cancers of the pancreas, breast, ovary, endometrium, and appendix, as well as gastric cancer and myeloma (Hartmaier et al. 2017). Treatment is based on tumor type as well as patient’s performance status, goals of care, and prior therapy.  However, because the prevalence of KRAS G12C-positive tumors is low, there is no specific guidance on treatment or management.

Although previously considered an "undruggable" cancer target, the landmark discovery of the switch II pocket within KRAS has enabled the development of covalent inhibitors specific for KRAS G12C (Ostrem et al. 2013). The clinical development of covalent inhibitors of KRAS G12C offers a potential therapeutic intervention to prevent or delay the progression of cancers that harbor a KRAS G12C mutation (McCormick 2019).

Early clinical data from covalent inhibitors specific for KRAS G12C demonstrate single-agent anti-tumor activity and potential to improve treatment options and outcomes for patients with advanced stage cancer that harbors the KRAS G12C mutation (Jänne et al. 2019; Hong et al. 2020).