MedPath

REGOMUNE : A phase I/II study of Regorafenib plus Avelumab in solid tumors

Phase 1/2
Active, not recruiting
Conditions
Adult patients with advanced or metastatic solid tumors
Registration Number
2023-509500-15-00
Lead Sponsor
Institut Bergonie
Brief Summary

Phase I : Primary objective of the phase I trial is to establish the recommended phase II dose (RP2D), the maximum tolerated dose (MTD) evaluated on the first cycle (D1 to D28),

the safety profile, and the dose limiting toxicities (DLT) of Regorafenib when prescribed in association with Avelumab (no dose escalation for Avelumab) in patients treated for advanced digestive solid tumors.

Phase II : To investigate the antitumor activity of Regorafenib when prescribed in association with Avelumab, independently for 19 cohorts of patients : Colorectal cancer not MSIH or MMR-deficient (Cohorts A and A’ with immune signature (based on low tumor-associated macrophages infiltrate level), GIST (Cohort B), Oesophageal or gastric carcinoma (Cohort C), Biliary tract cancer, hepatocellular carcinoma (Cohort D), Soft Tissue Sarcoma (STS – Cohort E), Radioiodine-Refractory Differentiated Thyroid Cancer (RR-DTC – Cohort F), Neuroendocrine gastroenteropancreatic tumors (GEP-NETs – Cohort G), Non-small cell lung cancer (NSCLC - Cohort H), Solid tumors (including Soft Tissue Sarcoma) with immune signature (TLS+) (Cohort I), urothelial cancer (Cohort J), HPV-associated cancer (Cohort K), triple negative breast cancer (L), TMB-high solid tumors (Cohort M), MSI-high solid tumors (Cohort N), non clear-cell renal carcinoma (Cohort O), malignant pleural mesothelioma (Cohort P), metastatic castration resistant prostate cancer (Cohort Q) with immune signature (TLS+) and neuroendocrine prostate cancer (Cohort R).

For cohorts A (Colorectal cancer not MSI-H or MMR-deficient [standard dose]), C (Oesophageal or gastric carcinoma), D (Biliary tract cancer, hepatocellular carcinoma), E (Soft Tissue Sarcoma [STS]), F (Radioiodine-Refractory Differentiated Thyroid Cancer), G (Neuroendocrine gastroenteropancreatic tumors):: antitumoral activity will be assessed in terms of objective response under treatment based on adapted RECIST 1.1 criteria after centralized radiological review (see endpoints section).

For cohorts B (GIST), H (NSCLC), I (Solid tumors -TLS+), M (TMB-high solid tumors), N (MSI-high solid tumors), O (non clear-cell renal carcinoma), P (malignant pleural mesothelioma) and R (neuroendocrine prostate cancer), antitumor activity will be assessed in terms of 6-month progression-free rate (6-month PFR) based on RECIST 1.1 criteria after centralized radiological review (see endpoints, section 9).

For cohort A’ with immune signature (based on low tumor-associated macrophages infiltrate level, antitumor activity will be assessed in terms of 4-month progressionfree rate (4-month PFR) based on RECIST 1.1 criteria after centralized radiological review (see endpoints, section 9).

For cohorts J, K and L, antitumor activity will be assessed in terms of disease control rate at 6-month (6-month DCR) based on RECIST 1.1 criteria after centralized radiological review (see endpoints, section 9).

For cohort Q (metastatic castration resistant prostate cancer with immune signature [TLS+]), antitumor activity will be assessed in terms of 6-month radiographic progression-free rate (6-month rPFR), after centralized radiological review, as per RECIST 1.1 for extra-skeletal lesions and PCWG3 for skeletal lesions criteria response criteria [88] (see endpoints, section 9).

Detailed Description

Not available

Recruitment & Eligibility

Status
Ongoing, recruitment ended
Sex
Not specified
Target Recruitment
827
Inclusion Criteria

Phase I - Histology : - Dose escalation part: histologically confirmed non MSI-H or MMR-deficient colorectal cancer, or GIST, or oesophageal or gastric carcinoma or hepatobiliary cancers.

Adequate hematological, renal, metabolic and hepatic functions: a. Hemoglobin ≥ 9 g/dl (patients may have received prior red blood cell [RBC] transfusion, if clinically indicated); absolute neutrophil count (ANC) ≥ 1.5 x 109/l and platelet count ≥ 100 x 109/l. b. Alkaline phosphatase (AP), alanine aminotransferase (ALT) and aspartate aminotransferase (ASP) ≤ 2.5 x upper limit of normality (ULN) (≤ 5 in case of extensive skeletal involvement and/or liver metastasis for AP and ≤ 5 x ULN in case of liver metastasis for AST and ALT). c. Total bilirubin ≤ 1.5 x ULN. d.Albumin ≥ 25g/l. e. Calculated creatinine clearance (CrCl) ≥ 30 ml/min (according to Cockroft and Gault formula). f. Creatine phosphokinase (CPK) ≤ 2.5 x ULN g. INR or PT ≤ 1.5 x ULN h. aPTT ≤ 1.5 X ULN. i. Lipase ≤ 1.5 X ULN j. Cohort specific criteria: Patients with hepatocellular carcinoma must have a correct hepatocellular function, id est Child-Pugh A. k. Cohort Q: serum testosterone < 50 ng/dL.

No prior or concurrent malignant disease diagnosed or treated in the last 3 years except for adequately treated in situ carcinoma of the cervix, basal or squamous skin cell carcinoma, or in situ transitional bladder cell carcinoma,

At least four weeks or 5 half-lives, whichever is shorter since last chemotherapy, immunotherapy or any other pharmacological treatment and/or radiotherapy,

Recovery to grade ≤ 1 from any adverse event (AE) derived from previous treatment, excluding alopecia of any grade and non-painful peripheral neuropathy grade ≤ 2 (according to the National Cancer Institute Common Terminology Criteria for Adverse Event (NCI-CTCAE, version 5.0)),

Women of childbearing potential must have a negative serum pregnancy test within 72 hours prior to receiving the first dose of study medication.

Both women and men must agree to use an highly effective method of contraception throughout the treatment period and for seven months (210 days) in WOCBP or four months (120 days) in men sexually active with WOCBP after discontinuation of treatment. Acceptable methods for contraception are described in section 7.4.1.

Voluntary signed and dated written informed consents prior to any specific study procedure,

Patients with a social security in compliance with the French law.

Documented disease progression (as per RECIST v1.1) before study entry. - For patient of cohorts E (STS) and cohort I (Solid tumors – TLS+) : this progression will be confirmed by central review on the basis of two CT scan or MRI obtained at less than 6 months in the period of 12 months prior to inclusion. - For patient of cohort F (RR-DTC) : this progression will be confirmed by central review on the basis of two CT scan or MRI obtained at less than 12 months prior to inclusion.

Cancelled (MSA6)

Phase II trials : histologically confirmed :  non MSI-H or MMR-deficient colorectal cancer (cohort A).  non MSI-H or MMR-deficient colorectal cancer with immune signature (cohort A’), i.e. low tumor-associated macrophages infiltrate level as determined by central review. Except if the low level of tumor-associated macrophages infiltrate level has been already confirmed by Biopathological platform at Bergonié Institute, the low level of tumor-associated macrophages infiltrate level should be confirmed by central review based on FFPE (Formalin-Fixed Paraffin-Embedded) tumor tissue sample (archived or newly obtained by biopsy for research purpose). Note that the level of tumor-associated macrophages infiltrate could be determined by central analysis if not available before.  or GIST (cohort B) : as recommended by INCa, patients must have diagnosis histologically confirmed by central review, except if it has been already confirmed by the RRePS Network.  or oesophageal or gastric carcinoma (cohort C)  or hepatobiliary cancers (cohort D)  or Soft Tissue Sarcoma (STS) (cohort E) : as recommended by INCa, patients must have diagnosis histologically confirmed by central review, except if it has been already confirmed by the RRePS Network  or Radioiodine-Refractory Differentiated Thyroid Cancer (RR-DTC) (cohort F)  or Neuroendocrine gastroenteropancreatic tumors (GEP-NETs) grade 2 and 3 (cohort G),  or Non-small cell lung cancer (cohort H),  or Solid tumors including soft-tissue sarcoma with immune signature (cohort I), i.e. presence of mature tertiary lymphoid structures (TLS). Except if presence of TLS has been already confirmed by Biopathological platform at Bergonié Institute, presence of TLS should be confirmed by central review based on FFPE (Formalin-Fixed Paraffin-Embedded) tumor tissue sample (archived or newly obtained by biopsy for research purpose). Note that the presence of TLS could be determined by central analysis if not available before.  or urothelial cancer (cohort J)  or HPV-associated cancer (cohort K) with molecular confirmation of p16 positive status.  or triple negative breast cancer (cohort L)  or TMB-high solid tumors (cohort M) with TMB-high status already known  or MSI-high solid tumors (cohort N) with MSI-high status already known  or Non clear-cell renal carcinoma (cohort O)  or Malignant pleural mesothelioma (cohort P)  or Metastatic castration resistant prostate cancer (cohort Q) with immune signature i.e., presence of mature tertiary lymphoid structures (TLS). TLS status determination can be centrally performed by Biopathological platform at Bergonié Institute or on each investigation site by a pathologist specifically trained by a representative of Biopathological platform at Bergonié Institute.  or neuroendocrine prostate cancer (cohort R) defined by the presence of 50% or more of neuroendocrine subtype in the tumor or the presence of 2 of the 3 subsequent genomic alterations: PTEN loss, RB1 mutation, TP53 mutation detected by NGS on tissue and/or blood samples.

For patients with non-small cell lung cancer (cohort H): - Subjects with histologically or cytologically confirmed diagnosis of nonsquamous NSCLC. - Documented disease progression based on radiographic imaging, during or after a maximum of 2 lines of systemic treatment for locally/regionally advanced recurrent, Stage IIIb/Stage IV or metastatic disease. Two components of treatment must have been received in the same line or as separate lines of therapy :  A maximum of 1 line of platinum-containing chemotherapy regimen in the metastatic setting, and  A maximum of 1 line of PD(L)1 mAb containing regimen, and  Patients must have received at least 4 months of PD(L1) mAb treatment. o No EGFR, ALK, ROS1 positive tumor mutations o Subjects with known BRAF molecular alterations must have had disease progression after receiving the locally available SoC treatment for the molecular alteration.

For patients with urothelial cancer (cohort J): - A maximum of 1 line of PD(L)1 mAb containing regimen, and - Patients must have received at least 4 months of PD(L1) mAb treatment.

For HPV-associated cancer (cohort K), TMB-high solid tumors (cohort M) MSIhigh solid tumors (cohort N), Non clear-cell renal carcinoma (cohort O): - A maximum of 1 line of PD(L)1 mAb containing regimen, and - Patients must have received at least 4 months of PD(L1) in the case they received this treatment

For malignant pleural mesothelioma (Cohort P): - A maximum of 1 line of PD(L)1/CTLA-4 mAb containing regimen, and - Patients must have received at least 4 months of PD(L1)/CTLA-4 mAb treatment in the case they received this treatment

For triple-negative breast cancer patients (Cohort L) : - A maximum of 1 line of PD(L)1 mAb containing regimen, and - Patients must have received at least 4 months of PD(L1) mAb treatment. Except if CPS<10, an anterior line of PD(L)1 mAb is not mandatory.

For TMB-High cancer patients (Cohort M): - TMB-High is defined as TMB score > 16 mutations /megabase on tissue or blood sample

For cohort Q: - progressive disease documented at inclusion either a PSA progression defined by a minimum of 2 rising PSA values of more than 2.0ng/mL with an interval of at least 1 week between the measurements OR bone disease progression as defined by PCWG3 criteria OR soft tissue disease progression as defined by RECIST 1.1 criteria . - Previous exposure to at least one new-hormone therapy (eg. abiraterone/enzalutamide/apalutamide/darolutamide). Patients may have received radionucleide-based therapies, or taxane based therapies.

For cohort R: previous exposure to a platinum-based chemotherapy (CT) or one new-hormone therapy (eg. abiraterone/enzalutamide/apalutamide/darolutamide). - If platinum-based CT was administered for localized disease, relapse and/or progression should occured within 12 months after the last CT administration to allow inclusion in the REGOMUNE study. Otherwise, platinum-based CT must be reintroduced before a potential inclusion in the REGOMUNE study. - If platinum-based CT was administered for advanced/metastatic disease, patient can be enrolled in the REGOMUNE study without delays.

Advanced non resectable / metastatic disease. For cohort Q, metastatic disease.

Patients for which either there is no further established therapy that is known to provide clinical benefit,

Age ≥ 18 years,

ECOG, Performance status ≤ 1,

Measurable disease according to RECIST v1.1,

Life expectancy > 3 months,

Except for cohorts F (RR-DTC), ≥ 1 previous line (s) of systemic therapy,

Exclusion Criteria

Previous treatment with Avelumab or Regorafenib,

Has a diagnosis of immunodeficiency or is receiving systemic steroid therapy or any other form of immunosuppressive therapy within 7 days prior to the first dose of trial treatment.

History of idiopathic pulmonary fibrosis (including pneumonitis), drug-induced pneumonitis, organizing pneumonia, or evidence of active pneumonitis on screening chest CT scan or interstitial lung disease with ongoing signs and symptoms at inclusion. History of radiation pneumonitis in the radiation field (fibrosis) is permitted.

Has known hepatitis B or hepatitis C, active and/or treated by antiviral therapy.

Has a known history of Human Immunodeficiency Virus (HIV) (HIV1/2 antibodies) or known acquired immunodeficiency syndrome (AIDS).

Spot urine must not show 1+ or more protein in urine or the patient will require a repeat urine analysis. If repeat urinalysis shows 1+ protein or more, a 24-hour urine collection will be required and must show total protein excretion <1000 mg/24 hours).

Major surgical procedure or significant traumatic injury within 28 days before start of study medication. Note: If a patient had major surgery, they must have recovered adequately from the toxicity and/or complications from the intervention prior to starting study treatment.

Non-healing wound, non-healing ulcer, or non-healing bone fracture requiring orthopedic treatment.

Patients with evidence or history of any bleeding diathesis, irrespective of severity.

Any hemorrhage or bleeding event ≥ CTCAE Grade 3 within 4 weeks prior to the start of study medication.

Arterial or venous thrombotic or embolic events such as cerebrovascular accident (including transient ischemic attacks), deep vein thrombosis or pulmonary embolism within 6 months before the start of study medication (except for adequately treated catheter-related venous thrombosis occurring more than one month before the start of study medication).

For cohorts A to G and A’: Has received prior therapy with an anti-PD-1, anti-PDL1, anti-PD-L2, anti-CD137, or anti-Cytotoxic T-lymphocyte-associated antigen- 4 (CTLA-4) antibody (including ipilimumab or any other antibody or drug specifically targeting T-cell co-stimulation or checkpoint pathways),

Ongoing infection > Grade 2 as per NCI CTCAE v5.0.

Uncontrolled hypertension (Systolic blood pressure > 140 mmHg or diastolic pressure > 90 mmHg) despite optimal medical management.

History of myocarditis or congestive heart failure ≥ New York Heart Association (NHYA) class 2.

Unstable angina (angina symptoms at rest), new-onset angina (begun within the last 3 months).

Myocardial infarction less than 6 months before start of study drug.

Uncontrolled cardiac arrhythmias.

Pregnant or breast-feeding patients.

Individuals deprived of liberty or placed under legal guardianship.

Prior organ transplantation, including allogeneic stem-cell transplantation.

Known alcohol or drug abuse.

Evidence of progressive or symptomatic or newly diagnosed central nervous system (CNS) or leptomeningeal metastases. Participants with previously treated brain metastases may participate provided they are stable (without evidence of progression by imaging for at least 4 weeks before the first dose of study treatment and any neurologic symptoms have returned to baseline), have no evidence of new or enlarging brain metastases confirmed by repeat imaging, and have not required steroids for at least 7 days before study treatment,

Vaccination within 4 weeks of the first dose of Avelumab and while on trial is prohibited except for administration of inactivated vaccines.

Patients with any condition that impairs their ability to swallow and retain tablets.

Other severe acute or chronic medical conditions including immune colitis, inflammatory bowel disease, immune pneumonitis, pulmonary fibrosis or psychiatric conditions including recent (within the past year) or active suicidal ideation or behavior; or laboratory abnormalities that may increase the risk associated with study participation or study treatment administration or may interfere with the interpretation of study results and, in the judgment of the investigator, would make the patient inappropriate for entry into this study.

Patient with anti-Vitamine K therapy.

Suspected or known intraabdominal fistula.

For cohort H (NSCLC): - Received > 2 prior lines of therapy for NSCLC, including subjects with BRAF molecular alterations, - Subjects with known EGFR/ALK/ROS1 molecular alterations are excluded from participation in this study.

Men or women of childbearing potential who are not using an effective method of contraception as previously described.

Participation to a study involving a medical or therapeutic intervention in the last 30 days.

Previous enrolment in the present study.

Patient unable to follow and comply with the study procedures because of any geographical, familial, social or psychological reasons.

Known hypersensitivity to any involved study drug or of its formulation components.

Active autoimmune disease that might deteriorate when receiving an immunostimulatory agent: a. Subjects with diabetes type I, vitiligo, psoriasis, hypo- or hyperthyroid disease not requiring immunosuppressive treatment are eligible. b. Subjects requiring hormone replacement with corticosteroids are eligible if the steroids are administered only for the purpose of hormonal replacement and at doses ≤ 10 mg or 10 mg equivalent prednisone per day. c. Administration of steroids through a route known to result in a minimal systemic exposure (topical, intranasal, intro-ocular, or inhalation) are acceptable.

Study & Design

Study Type
Interventional clinical trial of medicinal product
Study Design
Not specified
Primary Outcome Measures
NameTimeMethod
Phase I : Toxicity graded using the common toxicity criteria from the NC-CTCAE v5.

Phase I : Toxicity graded using the common toxicity criteria from the NC-CTCAE v5.

Phase I : Incidence rate of DLT at each dose level during the first 28 days.

Phase I : Incidence rate of DLT at each dose level during the first 28 days.

Phase II-Cohorts A,C,D,E,F,G: Antitumor activity will be assessed in terms of objective response under treatment based on adapted RECIST 1.1 criteria, and after centralized radiological review:

Phase II-Cohorts A,C,D,E,F,G: Antitumor activity will be assessed in terms of objective response under treatment based on adapted RECIST 1.1 criteria, and after centralized radiological review:

3a) Objective response under treatment is defined as patients with confirmed complete response (CR) or partial response, as per RECIST v1.1 criteria, observed during treatment with the investigational product(s).

3a) Objective response under treatment is defined as patients with confirmed complete response (CR) or partial response, as per RECIST v1.1 criteria, observed during treatment with the investigational product(s).

3b) As per RECIST v1.1 criteria, to be considered as “confirmed”, complete and partial responses will have to be confirmed at least 4 weeks later to ensure responses identified are not the result of measurement errors. If the new imaging to confirm response is not performed after 4 weeks, complete or partial responses will be considered as unconfirmed responses.

3b) As per RECIST v1.1 criteria, to be considered as “confirmed”, complete and partial responses will have to be confirmed at least 4 weeks later to ensure responses identified are not the result of measurement errors. If the new imaging to confirm response is not performed after 4 weeks, complete or partial responses will be considered as unconfirmed responses.

3c) Objective response rate (ORR) under treatment is defined as the proportion of patients with objective response (confirmed or unconfirmed) under treatment based on adapted RECIST 1.1.

3c) Objective response rate (ORR) under treatment is defined as the proportion of patients with objective response (confirmed or unconfirmed) under treatment based on adapted RECIST 1.1.

Phase II-Cohorts B,H,I,M,N,O,P,R: antitumor activity will be assessed in terms of 6-month progression-free rate (6- month PFR) based on RECIST 1.1 criteria after centralized radiological review. 6-month PFR is defined as the proportion of patients with progression-free status at 6 months. Progression-free status is defined as complete response (confirmed or unconfirmed), partial response (confirmed or unconfirmed) or stable disease more than 24 weeks as defined as per RECIST v1.1 criteria.

Phase II-Cohorts B,H,I,M,N,O,P,R: antitumor activity will be assessed in terms of 6-month progression-free rate (6- month PFR) based on RECIST 1.1 criteria after centralized radiological review. 6-month PFR is defined as the proportion of patients with progression-free status at 6 months. Progression-free status is defined as complete response (confirmed or unconfirmed), partial response (confirmed or unconfirmed) or stable disease more than 24 weeks as defined as per RECIST v1.1 criteria.

Phase II-Cohort A': antitumor activity will be assessed in terms of 4- month progression-free rate (4-month PFR) based on RECIST 1.1 criteria after centralized radiological review. 4-month PFR is defined as the proportion of patients with progression-free status at 4 months. Progression-free status is defined as complete response (confirmed or unconfirmed), partial response (confirmed or unconfirmed) or stable disease more than 24 weeks as defined as per RECIST v1.1 criteria.

Phase II-Cohort A': antitumor activity will be assessed in terms of 4- month progression-free rate (4-month PFR) based on RECIST 1.1 criteria after centralized radiological review. 4-month PFR is defined as the proportion of patients with progression-free status at 4 months. Progression-free status is defined as complete response (confirmed or unconfirmed), partial response (confirmed or unconfirmed) or stable disease more than 24 weeks as defined as per RECIST v1.1 criteria.

Phase II-Cohorts J,K,L:antitumor activity will be assessed in terms of disease control rate at 6-month based on RECIST 1.1 criteria after centralized radiological review. 6-month DCR rate is defined as the proportion of participants with confirmed complete response, unconfirmed complete response, confirmed partial response or unconfirmed partial response within 24 weeks of treatment onset (while treated with the investigational product), or stable disease at 6 months, as per adapted RECIST v1.1.

Phase II-Cohorts J,K,L:antitumor activity will be assessed in terms of disease control rate at 6-month based on RECIST 1.1 criteria after centralized radiological review. 6-month DCR rate is defined as the proportion of participants with confirmed complete response, unconfirmed complete response, confirmed partial response or unconfirmed partial response within 24 weeks of treatment onset (while treated with the investigational product), or stable disease at 6 months, as per adapted RECIST v1.1.

Phase II-Cohort Q: antitumor activity will be assessed in terms of 6-month radiographic progression-free rate (6-month rPFR) after centralized radiological review, as per RECIST 1.1 for extra-skeletal lesions and PCWG3 for skeletal lesions criteria response criteria [88]. 6-month rPFR is defined as the proportion of patients with radiographic progression-free status at 6 months. Radiographic progression is defined as per RECIST/PCWG3 criteria as soft-tissue and/or bone progression as follows :

Phase II-Cohort Q: antitumor activity will be assessed in terms of 6-month radiographic progression-free rate (6-month rPFR) after centralized radiological review, as per RECIST 1.1 for extra-skeletal lesions and PCWG3 for skeletal lesions criteria response criteria [88]. 6-month rPFR is defined as the proportion of patients with radiographic progression-free status at 6 months. Radiographic progression is defined as per RECIST/PCWG3 criteria as soft-tissue and/or bone progression as follows :

10a) Soft tissue: Radiographic progression is defined as an increase in measurable disease according to Response Evaluation Criteria in Solid Tumors (RECIST v1.1).

10a) Soft tissue: Radiographic progression is defined as an increase in measurable disease according to Response Evaluation Criteria in Solid Tumors (RECIST v1.1).

10b) Bone: Bone progression is defined by the occurrence of at least 2 new lesions.

10b) Bone: Bone progression is defined by the occurrence of at least 2 new lesions.

Secondary Outcome Measures
NameTimeMethod
Phase I secondary endpoints : Preliminary signs of antitumor activity in terms of:

Phase I secondary endpoints : Preliminary signs of antitumor activity in terms of:

1a) Best overall response defined as the best response recorded from the start of the study treatment until the end of treatment taking into account any requirement for confirmation as per RECIST v1.1 criteria.

1a) Best overall response defined as the best response recorded from the start of the study treatment until the end of treatment taking into account any requirement for confirmation as per RECIST v1.1 criteria.

1b) Objective response rate (ORR) defined as the proportion of patients with complete response or partial response, as per RECIST 1. ORR under treatment and 6-month ORR will be reported.

1b) Objective response rate (ORR) defined as the proportion of patients with complete response or partial response, as per RECIST 1. ORR under treatment and 6-month ORR will be reported.

1c) Progression-free rate (PFR) at 6 months defined as the proportion of patients with complete response, partial response or stable disease more than 24 weeks as per RECIST v1.1 criteria.

1c) Progression-free rate (PFR) at 6 months defined as the proportion of patients with complete response, partial response or stable disease more than 24 weeks as per RECIST v1.1 criteria.

1d) Progression-free survival (PFS) defined as the time from study treatment initiation to the first occurrence of disease progression or death (of any cause), whichever occurs first. 1-year PFS rate and median PFS will be reported.

1d) Progression-free survival (PFS) defined as the time from study treatment initiation to the first occurrence of disease progression or death (of any cause), whichever occurs first. 1-year PFS rate and median PFS will be reported.

1e) Overall Survival (OS) defined as the time from study treatment initiation to death (of any cause). 1-year OS rate and median OS will be reported.

1e) Overall Survival (OS) defined as the time from study treatment initiation to death (of any cause). 1-year OS rate and median OS will be reported.

1f) Growth modulation index (GMI): GMI will be defined for each patient as the ratio of its PFS on Regorafenib + Avelumab treatment to its PFS on the previous line of therapy. This method accounts for inter-patient variability, the patient serving as his/her own control and implies by the natural history of the disease that the PFS tends to become shorter in successive lines of therapy. It is thought that an anti-cancer agent should be considered effective if the GMI is greater than 1.3.

1f) Growth modulation index (GMI): GMI will be defined for each patient as the ratio of its PFS on Regorafenib + Avelumab treatment to its PFS on the previous line of therapy. This method accounts for inter-patient variability, the patient serving as his/her own control and implies by the natural history of the disease that the PFS tends to become shorter in successive lines of therapy. It is thought that an anti-cancer agent should be considered effective if the GMI is greater than 1.3.

Phase I secondary endpoints :PK measurements expressed as AUC, half-life and concentration peak for Regorafenib

Phase I secondary endpoints :PK measurements expressed as AUC, half-life and concentration peak for Regorafenib

Phase I secondary endpoints : Pharmacodynamic activity: Predictive biomarkers analysis and pharmacodynamic (PD)/mechanism of action (MOA) in blood (levels of angiogenic and immunologic biomarkers in blood at baseline and different study time points), potentially including but not limited to:

Phase I secondary endpoints : Pharmacodynamic activity: Predictive biomarkers analysis and pharmacodynamic (PD)/mechanism of action (MOA) in blood (levels of angiogenic and immunologic biomarkers in blood at baseline and different study time points), potentially including but not limited to:

9a) Serum/plasma cytokines levels (ELISA)

9a) Serum/plasma cytokines levels (ELISA)

9b) Treg, CD4+, CD8+ and DR lymphocytes subpopulations (flow cytometry)

9b) Treg, CD4+, CD8+ and DR lymphocytes subpopulations (flow cytometry)

9c) Archived tumor tissue will be collected for assessment of tumor VEGFR, PDGFR, HIF1alpha expression and lymphocytes, TAM and MDSC tumor infiltrates (IHC)

9c) Archived tumor tissue will be collected for assessment of tumor VEGFR, PDGFR, HIF1alpha expression and lymphocytes, TAM and MDSC tumor infiltrates (IHC)

9d) In additional, for all patients, optional biopsy at baseline and after 4 weeks of treatment will be proposed for mechanisms of action documentation: tumor VEGFR, PDGFR, HIF1alpha expression as well as PD-L1/PD1, lymphocytes, TAM, MDSC tumor infiltrates (IHC) and mutational burden.

9d) In additional, for all patients, optional biopsy at baseline and after 4 weeks of treatment will be proposed for mechanisms of action documentation: tumor VEGFR, PDGFR, HIF1alpha expression as well as PD-L1/PD1, lymphocytes, TAM, MDSC tumor infiltrates (IHC) and mutational burden.

Phase II secondary endpoints [All cohorts, except cohort Q]: Best overall response is defined as the best response across all time points (RECIST v1.1). Following RECIST v1.1 recommendations:

Phase II secondary endpoints [All cohorts, except cohort Q]: Best overall response is defined as the best response across all time points (RECIST v1.1). Following RECIST v1.1 recommendations:

14a) The best overall response is determined once all the data for the patient is known.

14a) The best overall response is determined once all the data for the patient is known.

14b) The best overall response will be classified as confirmed complete response (CR), unconfirmed complete response (CRu), confirmed partial response (PR), unconfirmed partial response (PRu), stable disease or progressive disease, as per RECIST v1.1 criteria.

14b) The best overall response will be classified as confirmed complete response (CR), unconfirmed complete response (CRu), confirmed partial response (PR), unconfirmed partial response (PRu), stable disease or progressive disease, as per RECIST v1.1 criteria.

14c) As per RECIST v1.1 criteria, to be considered as “confirmed”, complete and partial responses will have to be confirmed at least 4 weeks later to ensure responses identified are not the result of measurement errors.

14c) As per RECIST v1.1 criteria, to be considered as “confirmed”, complete and partial responses will have to be confirmed at least 4 weeks later to ensure responses identified are not the result of measurement errors.

Phase II [All cohorts, except cohort Q]: Objective response rate at 6 months (6-month ORR) is defined as the proportion of patients with objective response (confirmed or unconfirmed) at 6 months.

Phase II [All cohorts, except cohort Q]: Objective response rate at 6 months (6-month ORR) is defined as the proportion of patients with objective response (confirmed or unconfirmed) at 6 months.

Phase II [All cohorts, except cohort Q]: Progression-free status is defined as complete response (confirmed or unconfirmed), partial response (confirmed or unconfirmed) or stable disease more than 24 weeks as defined as per RECIST v1.1 criteria (appendix 3). 6-month progression-free rate (6-month PFR) is defined as the proportion of patients with progression-free status at 6 months. 4- month progression-free rate is defined as the proportion of patients with progression-free status at 4 months.

Phase II [All cohorts, except cohort Q]: Progression-free status is defined as complete response (confirmed or unconfirmed), partial response (confirmed or unconfirmed) or stable disease more than 24 weeks as defined as per RECIST v1.1 criteria (appendix 3). 6-month progression-free rate (6-month PFR) is defined as the proportion of patients with progression-free status at 6 months. 4- month progression-free rate is defined as the proportion of patients with progression-free status at 4 months.

Phase II Cohort Q : PSA response rate is defined as the percentage of patients reaching a PSA decline from baseline ≥ 50% with PSA confirmation ≥ 3 weeks after the first documented reduction in PSA of ≥ 50%. The PSA response rate will be calculated as the proportion of participants with a PSA response in the PSA response-evaluable population.

Phase II Cohort Q : PSA response rate is defined as the percentage of patients reaching a PSA decline from baseline ≥ 50% with PSA confirmation ≥ 3 weeks after the first documented reduction in PSA of ≥ 50%. The PSA response rate will be calculated as the proportion of participants with a PSA response in the PSA response-evaluable population.

Phase II Cohort Q : The 1-year PSA response rate will be calculated as the proportion of participants with a PSA response at one year. after the first documented reduction in PSA

Phase II Cohort Q : The 1-year PSA response rate will be calculated as the proportion of participants with a PSA response at one year. after the first documented reduction in PSA

Phase II Cohort Q : PSA-Progression Free Survival (PSA-PFS) is defined as the time from treatment initiation until first evidence of PSA progression or until death from any cause, whichever comes first. PSA progression is defined by the criteria of the Prostate Cancer Clinical Trials Working Group (PCWG 3) as follows:

Phase II Cohort Q : PSA-Progression Free Survival (PSA-PFS) is defined as the time from treatment initiation until first evidence of PSA progression or until death from any cause, whichever comes first. PSA progression is defined by the criteria of the Prostate Cancer Clinical Trials Working Group (PCWG 3) as follows:

22a) For patients with a PSA decrease observed after baseline: PSA progression is defined as the first PSA increase that is ≥ 25% and ≥ 2 ng/mL above the nadir, and which is confirmed by a second value ≥ 1 week later (i.e., a confirmed rising trend). The date of progression corresponds to the date of the first PSA increase that is ≥ 25% and ≥ 2 ng/mL above the nadir.

22a) For patients with a PSA decrease observed after baseline: PSA progression is defined as the first PSA increase that is ≥ 25% and ≥ 2 ng/mL above the nadir, and which is confirmed by a second value ≥ 1 week later (i.e., a confirmed rising trend). The date of progression corresponds to the date of the first PSA increase that is ≥ 25% and ≥ 2 ng/mL above the nadir.

22b) For patients without a PSA decrease observed after baseline: PSA progression is defined as PSA progression ≥ 25% increase and ≥ 2 ng/mL increase from baseline beyond 12 weeks.

22b) For patients without a PSA decrease observed after baseline: PSA progression is defined as PSA progression ≥ 25% increase and ≥ 2 ng/mL increase from baseline beyond 12 weeks.

Phase II : Growth modulation index (GMI): GMI is defined for each patient as the ratio of its PFS on Regorafenib + Avelumab treatment to its PFS on the previous line of therapy. This method accounts for inter-patient variability, the patient serving as his/her own control and implies by the natural history of the disease that the PFS tends to become shorter in successive lines of therapy. It is thought that an anticancer agent should be considered effective if the GMI is greater than 1.3.

Phase II : Growth modulation index (GMI): GMI is defined for each patient as the ratio of its PFS on Regorafenib + Avelumab treatment to its PFS on the previous line of therapy. This method accounts for inter-patient variability, the patient serving as his/her own control and implies by the natural history of the disease that the PFS tends to become shorter in successive lines of therapy. It is thought that an anticancer agent should be considered effective if the GMI is greater than 1.3.

Phase II : Progression-free survival (PFS) is defined as the time from study treatment initiation to the first occurrence of disease progression or death (of any cause), whichever occurs first. 1-year PFS rate and median PFS will be reported.

Phase II : Progression-free survival (PFS) is defined as the time from study treatment initiation to the first occurrence of disease progression or death (of any cause), whichever occurs first. 1-year PFS rate and median PFS will be reported.

Phase II : Overall Survival (OS) is defined as the time from study treatment initiation to death (of any cause). 1-year OS rate and median OS will be reported.

Phase II : Overall Survival (OS) is defined as the time from study treatment initiation to death (of any cause). 1-year OS rate and median OS will be reported.

Phase II : Safety profile of the association Regorafenib + Avelumab: Toxicity will be graded using the common toxicity criteria from the NCI v5.0.

Phase II : Safety profile of the association Regorafenib + Avelumab: Toxicity will be graded using the common toxicity criteria from the NCI v5.0.

Phase II : Pharmacodynamic activity: o archived tumor tissue will be collected for assessment of the tumor microenvironment. o to perform integrative assessment of biomarkers of efficacy (genetic, metabolomics profiling in blood/tissue at baseline and different study time points) and its prognostic value on efficacy.

Phase II : Pharmacodynamic activity: o archived tumor tissue will be collected for assessment of the tumor microenvironment. o to perform integrative assessment of biomarkers of efficacy (genetic, metabolomics profiling in blood/tissue at baseline and different study time points) and its prognostic value on efficacy.

Phase II Cohort B : To evaluate the antitumor activity based on Choi criteria in terms of non-progression at 6 months.

Phase II Cohort B : To evaluate the antitumor activity based on Choi criteria in terms of non-progression at 6 months.

Trial Locations

Locations (7)

Institut Gustave Roussy

🇫🇷

Villejuif Cedex, France

Institut Bergonie

🇫🇷

Bordeaux, France

Oncopole Claudius Regaud

🇫🇷

Toulouse, France

Institut Regional Du Cancer De Montpellier

🇫🇷

Montpellier Cedex 5, France

Centre Leon Berard

🇫🇷

Lyon, France

Centre Hospitalier Regional Et Universitaire De Brest

🇫🇷

Brest, France

Institut Curie

🇫🇷

Paris, France

Institut Gustave Roussy
🇫🇷Villejuif Cedex, France
Rastislav BAHLEDA
Site contact
0142114316
rastislav.bahleda@gustaveroussy.fr
© Copyright 2025. All Rights Reserved by MedPath