Clinical Trials Logo

Clinical Trial Details — Status: Recruiting

Administrative data

NCT number NCT06154291
Other study ID # XT 23-01
Secondary ID 2023-505266-29-0
Status Recruiting
Phase Phase 1/Phase 2
First received
Last updated
Start date November 14, 2023
Est. completion date November 2027

Study information

Verified date January 2024
Source Xenothera SAS
Contact Françoise SHNEIKER, MD
Phone +33652720301
Email francoise.shneiker@xenothera.com
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This is a two-stage trial consisting of a Part I, dose escalation and dose-finding component to establish the Maximal Tolerated Dose (MTD), if any, and Recommended Part 2 Dose (RP2D) of XON7, followed by a Part II component to investigate anti-tumors efficacy in selected solid tumor types and to further evaluate safety and tolerability of XON7 at RP2D.


Recruitment information / eligibility

Status Recruiting
Enrollment 255
Est. completion date November 2027
Est. primary completion date December 2026
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: 1. Provide signed, written informed consent. 2. Male and female participant, age = 18 years old (at the time consent is obtained) 3. Solid tumors indications: - Participant in phase I, must have a histologically or cytologically confirmed advanced or metastatic solid tumors for which no effective standard therapy is available. All tumor types except glioblastoma, could be included. - Participant in phase II, must have histologically or cytologically confirmed advanced or metastatic solid tumors of the following: NSCLC, gastro-esophageal adenocarcinoma, CRC, pancreatic cancer, Sarcoma, TNBC, or ovarian cancer. 4. Line of treatment: Participant must have solid tumors progressing after = 4 lines of standard appropriate anticancer therapies for the specific tumor type, or for which the patient is ineligible. Participants whose cancers harbor molecular alterations for which targeted therapy is standard of care should have received health authority-approved appropriate targeted therapy for their tumor types before enrollment. 5. Measurable disease per RECIST version 1.1 - v5 6. (ECOG) performance status (PS) 0-1 7. Life expectancy of at least 12 weeks. 8. Adequate organ function 9. QT duration corrected for heart rate by Fridericia's formula (QTcF) <450 msec or QTcF <480 msec for participants with bundle branch block. 10. In France, a participant will be eligible for inclusion in this trial only if either affiliated to or a beneficiary of a social security category. 11. Female participant who are not of child-bearing potential, and female participants of child-bearing potential who have a negative serum pregnancy test within 7 days prior to initial trial treatment. Female participants of child-bearing potential, and all male partners must consent to use a medically acceptable method of contraception throughout the trial period and for at least 60 days after the last dose of XON7. A barrier method of contraception must be included. 12. Male participant willing to use adequate contraceptive measures throughout the trial period and for at least 60 days after the last dose of trial intervention. 13. For phase II, participant in pharmacodynamics cohort must provide biopsy of a tumor lesion not previously irradiated during the screening period and must agree to provide at least one additional on-treatment biopsy between day 36 and 42 after trial intervention administration. 14. For phase II, participant in pharmacodynamics cohort must have accessible tumor tissue available for fresh biopsy except for ovarian cancer and sarcoma. Exclusion Criteria: 1. A participant who has received more than 4 prior lines of therapy for advanced or metastatic disease. 2. A participant who has had a prior anti-cancer mAb within 3 weeks prior to trial Day 1 or who has not recovered (i.e., = Grade 1 or at baseline) from adverse events due to agents administered more than 4 weeks earlier prior to trial Day 1. 3. A participant who has had prior chemotherapy, targeted small molecule therapy, or radiation therapy within 2 weeks prior to trial Day 1 or who have not recovered (i.e., = Grade 1 or at baseline) from adverse events due to a previously administered agent (Except alopecia, hearing loss, grade 2 neuropathy or endocrinopathy managed with replacement therapy). 4. A participant with =Grade 3 toxicity related to prior immunotherapy leading to treatment discontinuation. 5. A participant whose toxicity related to prior treatment has not resolved to Grade 1 (except alopecia, hearing loss, grade 2 neuropathy or endocrinopathy managed with replacement therapy). 6. A participant who has received major surgery 2 weeks before the first dose of trial treatment or has not recovered adequately from the toxicity and/or complications from any surgery (major or minor) before initiating trial treatment. 7. Concomitant use of another experimental drug, or wash-out period of at least 5 half-lives for a previous experimental drug not completed before start of trial intervention 8. Participant treated with drugs known to prolong the QT interval 9. Participant with carcinomatous meningitis. 10. Central nervous system (CNS) metastases, with the exception of individuals who have been previously treated CNS metastases, are asymptomatic, and have had no requirement for steroids for 3 weeks prior to first dose of trial drug. 11. Malignancies other than disease under trial within 3 years prior to first dose of trial intervention. 12. History of autoimmune disease 13. Active or uncontrolled infections requiring systemic treatment (known human immunodeficiency virus infection, or positive test for hepatitis B surface antigen or hepatitis C). 14. Any severe and/or uncontrolled medical conditions or other conditions that could affect their participation in the trial such as history or evidence of cardiovascular risk including any of the following: - Recent (within the past 6 months) history of serious uncontrolled cardiac arrhythmia or clinically significant ECG abnormalities including second degree (Type II) or third-degree atrioventricular block. - Documented cardiomyopathy, myocardial infarction, acute coronary syndromes (including unstable angina pectoris), coronary angioplasty, stenting, or bypass grafting within the past 6 months before enrollment. - Documented congestive heart failure (Class II, III, or IV) as defined by the New York Heart Association functional classification system (NYHA, 1994). 15. Prior allogeneic or autologous bone marrow transplantation or other solid organ transplantation. 16. Current active liver or biliary disease (Except for Gilbert's syndrome or asymptomatic gallstones, liver metastases, or otherwise stable chronic liver disease per investigator assessment). 17. Concurrent medical condition requiring the use of systemic immunosuppressive medications within 28 days before the first dose of trial treatment. 18. Recent history (within the past 6 months) of acute diverticulitis, inflammatory bowel disease, intra-abdominal abscess, or gastrointestinal obstruction. 19. Current or history of idiopathic pulmonary fibrosis, interstitial lung disease, or organizing pneumonia. Note: post-radiation changes in the lung related to prior radiotherapy and/or asymptomatic radiation-induced pneumonitis not requiring treatment may be permitted if agreed by the investigator and Medical Monitor. 20. History of (non-infectious) pneumonitis that required steroids or current pneumonitis. 21. Recent history (within 6 months) of uncontrolled symptomatic ascites or pleural effusions. 22. Participant who has received transfusion of blood products (including platelets or red blood cells) or administration of colony stimulating factors (including granulocyte colony- stimulating factor [G-CSF], granulocyte-macrophage colony-stimulating factor [GM- CSF], recombinant erythropoietin) within 2 weeks before the first dose of trial intervention. 23. Known, current drug or alcohol abuse. 24. Female participant who is pregnant or lactating. 25. Psychological, familial, sociological, or geographical conditions that do not permit compliance with the protocol. 26. Inability or unwillingness to comply with trial and/or follow-up procedures outlined in the protocol. 27. For France, patients under legal protection (safeguard, guardianship, curatorship)

Study Design


Intervention

Drug:
XON7
The trial intervention (XON7) is a glyco-humanized polyclonal antibody drug which is formulated for intravenous (IV) administration

Locations

Country Name City State
Belgium Institut Jules Bordet Anderlecht
France Centre Léon Bérard Lyon
France Hôpital Foch Suresnes
France IUCT-Oncopole Toulouse

Sponsors (1)

Lead Sponsor Collaborator
Xenothera SAS

Countries where clinical trial is conducted

Belgium,  France, 

Outcome

Type Measure Description Time frame Safety issue
Primary Dose Escalation part: Dose Limiting Toxicities (DLTs) Investigator defined DLT during first treatment cycle At the end of Cycle 1 (28 days)
Primary Dose Escalation part: treatment emergent adverse events (TEAEs) An overall summary of AE occurrences with onset during the first treatment cycle will be presented; this will include the following AE attributes:
Preferred term,
Maximum CTCAE grade,
Outcome,
Time to first occurrence [days].
At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Leucocytes count Incidence and magnitude of clinically significant changes in Leucocytes Count (G/L) At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Red Blood Cells Count Incidence and magnitude of clinically significant changes in Red Blood Cells Count (T/L) At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Hemoglobin Incidence and magnitude of clinically significant changes in Hemoglobin (g/dL). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Hematocrit Incidence and magnitude of clinically significant changes in Hematocrit (%). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Absolute Neutrophil Count Incidence and magnitude of clinically significant changes in Absolute Neutrophil Count (G/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Absolute Eosinophil Count Incidence and magnitude of clinically significant changes in Absolute Eosinophil Count (G/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Absolute Basophil Count Incidence and magnitude of clinically significant changes in Absolute Basophil Count (G/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Absolute Lymphocytes Count Incidence and magnitude of clinically significant changes in Absolute Lymphocytes Count (G/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Absolute Monocytes Count Incidence and magnitude of clinically significant changes in Absolute Monocytes Count (G/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Platelet Count Incidence and magnitude of clinically significant changes in Platelet Count (G/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Albumin Incidence and magnitude of clinically significant changes in Albumin (g/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Bicarbonate Incidence and magnitude of clinically significant changes in Bicarbonate (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Total Bilirubin Incidence and magnitude of clinically significant changes in Total Bilirubin (µmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Calcium Incidence and magnitude of clinically significant changes in Calcium (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Urea Incidence and magnitude of clinically significant changes in Urea (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Chloride Incidence and magnitude of clinically significant changes in Chloride (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Creatinine Incidence and magnitude of clinically significant changes in Creatinine (µmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Creatinine Clearance Incidence and magnitude of clinically significant changes in Creatinine Clearance (mL/min). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Glucose Incidence and magnitude of clinically significant changes in Glucose (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Magnesium Incidence and magnitude of clinically significant changes in Magnesium (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Phosphate Incidence and magnitude of clinically significant changes in Phosphate (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Potassium Incidence and magnitude of clinically significant changes in Potassium (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Sodium Incidence and magnitude of clinically significant changes in Sodium (mmol/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Total Protein Incidence and magnitude of clinically significant changes in Total Protein (g/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Alanine aminotransferase (ALT) Incidence and magnitude of clinically significant changes in Alanine aminotransferase (ALT) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Aspartate aminotransferase (AST) Incidence and magnitude of clinically significant changes in Aspartate aminotransferase (AST) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Gamma-glutamyl transférase (GGT) Incidence and magnitude of clinically significant changes in Gamma-glutamyl transférase (GGT) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Alkaline Phosphatase (ALP) Incidence and magnitude of clinically significant changes in Alkaline Phosphatase (ALP) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Lactate dehydrogenase (LDH) Incidence and magnitude of clinically significant changes in Lactate dehydrogenase (LDH) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Amylase Incidence and magnitude of clinically significant changes in Amylase (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Lipase Incidence and magnitude of clinically significant changes in Lipase (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Total Protein Creatine Kinase (CK) Incidence and magnitude of clinically significant changes in Total Protein Creatine Kinase (CK) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Creatine kinase - cardiac muscle isoenzyme (CK-MB) Incidence and magnitude of clinically significant changes in Creatine kinase - cardiac muscle isoenzyme (CK-MB) (U/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Troponin T Incidence and magnitude of clinically significant changes in Troponin T (ng/L). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Prothrombin Time (PT) Incidence and magnitude of clinically significant changes in Prothrombin Time (PT) (Sec). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in INR (if under VKA Therapy) Incidence and magnitude of clinically significant changes in INR (if under VKA Therapy) At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant changes in Activated Partial Thromboplastin Time (aPTT) Incidence and magnitude of clinically significant changes in Activated Partial Thromboplastin Time (aPTT) (Sec). At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant findings in blood pressure (BP) Incidence and severity of clinically significant findings in blood pressure: Significant blood pressure increase will be defined as BP >150/100 mmHg in a subject without a history of hypertension or increased >20 mmHg (diastolic) from baseline measurement in a subject with a previous history of hypertension. At the end of Cycle 1 (28 days)
Primary Dose Escalation part: clinically significant findings in electrocardiogram (ECGs) Incidence of clinically significant findings in Electrocardiogram (ECG): Significant QTc prolongation will be defined as an interval =500 msec or an interval which increases by =60 msec over baseline At the end of Cycle 1 (28 days)
Primary Expansion part: Anti-tumors efficacy Objective response rate (ORR): defined as the proportion of participants with a confirmed complete response [CR] or confirmed partial response [PR] assessed by investigators according to RECIST v1.1. Within 3 months after XON7 initiation
Secondary Pharmacokinetics (PK) of XON7 (Part 1): Cmax XON7 peak plasma concentration (Cmax) in plasma Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): Cmax XON7 peak plasma concentration (Cmax) in plasma Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): Tmax Time to peak drug concentration in plasma (Tmax) Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): Tmax Time to peak drug concentration in plasma (Tmax) Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): AUC Area under the plasma concentration versus time curve (AUC). AUC24hours; AUC0-14days and AUC15-28days will be assessed Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): AUC Area under the plasma concentration versus time curve (AUC). AUC24hours; AUC0-14days and AUC15-28days will be assessed Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): Ctrough Trough concentration (Ctrough) is the concentration reached by XON7 immediately before the next dose is administered Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): Ctrough Trough concentration (Ctrough) is the concentration reached by XON7 immediately before the next dose is administered Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): Cmin Cmin for the minimum blood plasma concentration reached by XON7 during the time interval between administration of two doses Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): Cmin Cmin for the minimum blood plasma concentration reached by XON7 during the time interval between administration of two doses Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): T1/2 Half-life (T1/2) refers to the time required for plasma concentration of XON7 to decrease by 50% Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): T1/2 Half-life (T1/2) refers to the time required for plasma concentration of XON7 to decrease by 50% Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): CL Clearance (CL) is the volume of blood or plasma cleared of XON7 from the body per unit of time Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): CL Clearance (CL) is the volume of blood or plasma cleared of XON7 from the body per unit of time Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 1): Vd Volume of distribution (Vd) is defined as the total amount of XON7 in the body divided by its concentration in plasma Cycle 1-Day 1 Predose and Postdose: 5 minutes; 1; 2; 4; 8; 24; 72 or 96; 144 hours after the end of infusion. Cycle 1 Day 15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 7 Day 1: Predose and 5 minutes after the end of infusion
Secondary Pharmacokinetics (PK) of XON7 (Part 2): Vd Volume of distribution (Vd) is defined as the total amount of XON7 in the body divided by its concentration in plasma Cycle 1-Day 1; Cycle 2-Day 1; Cycle 3-Day 1; Cycle 6-Day 1; Cycle 9-Day1 and Cycle 12-Day 1: Predose and 5 minutes after the end of infusion
Secondary Host immunogenicity to XON7 (Part 1 and part 2) Number of participants who develop detectable anti-drug antibodies Cycle 1-Day 1; Cycle 1-Day4; Cycle 1-Day7; Cycle 1-Day15; Cycle 2 and additional cycles-Day1; 30 and 60 days after the last dose of XON7
Secondary Host immunogenicity to XON7 (Part 1 and part 2) Percentage of participants who develop detectable anti-drug antibodies Cycle 1-Day 1; Cycle 1-Day4; Cycle 1-Day7; Cycle 1-Day15; Cycle 2 and additional cycles-Day1; 30 and 60 days after the last dose of XON7
Secondary Further assess anti-tumor efficacy (Part 1): ORR Objective response rate (ORR): defined as the proportion of participants with a confirmed complete response [CR] or confirmed partial response [PR] assessed by investigators according to RECIST v1.1. within 3 months after XON7 initiation Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Further assess anti-tumor efficacy (Part 2): CBR Clinical Benefit Rate (CBR) defined as the proportion of participants who achieve CR, PR, and durable SD [SD=24 weeks] assessed by investigators according to the RECIST criteria v 1.1. Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Further assess anti-tumor efficacy (Part 1 and part 2): DCR Disease control rate (DCR): defined as the proportion of participants who achieve confirmed complete response [CR], confirmed partial response [PR] or stable disease [SD] assessed by investigators according to the RECIST criteria version 1.1. Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Further assess anti-tumor efficacy (Part 1 and part 2): DoR Duration of response (DoR): defined as the time interval between the first confirmed objective response (CR or PR per RECIST 1.1 by investigators) and the first occurrence of objective tumor progression (Progressive disease (PD) per RECIST 1.1 by investigators) or death from any cause. Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Further assess anti-tumor efficacy (Part 2): TTR Time to response (TTR): defined as the time from the date of XON7 initiation to first confirmed objective response (CR or PR per RECIST 1.1 by investigators).. Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Further assess anti-tumor efficacy (Part 1 and part 2): PFS Progression-free survival (PFS): defined as the time from the date of XON7 initiation to the date of first documented progression (RECIST 1.1 by investigators) or death. Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Further assess anti-tumor efficacy (Part 1 and part 2): OS Overall survival (OS): defined as the time interval between the date of XON7 initiation and the date of death due to any cause. Tumor imaging (computed tomography [CT]) will be performed within 28 days prior to enrollment, and while on study approximately every 8 weeks during the treatment period. During the survival follow-up, results of CT only performed in current practice
Secondary Expansion part: treatment emergent adverse events (TEAEs) An overall summary of AE occurrences with onset during the first treatment cycle will be presented; this will include the following AE attributes:
Preferred term,
Maximum CTCAE grade,
Outcome,
Time to first occurrence [days].
Participants will be assessed for AEs and SAEs beginning immediately after the first dose of investigational drug and continuing through to follow-up which is 60 ± 5 days of last dose of investigational drug.
Secondary Dose Escalation part: clinically significant findings in blood pressure (BP) Incidence and severity of clinically significant findings in blood pressure: Significant blood pressure increase will be defined as BP >150/100 mmHg in a subject without a history of hypertension or increased >20 mmHg (diastolic) from baseline measurement in a subject with a previous history of hypertension. Before and immediately after the first dose of investigational drug and continuing through to follow-up which is 60 ± 5 days of last dose of investigational drug
Secondary Dose Escalation part: clinically significant findings in electrocardiogram (ECGs) Incidence of clinically significant findings in Electrocardiogram (ECG): Significant QTc prolongation will be defined as an interval =500 msec or an interval which increases by =60 msec over baseline Before and immediately after the first dose of investigational drug and continuing through to follow-up which is 60 ± 5 days of last dose of investigational drug
Secondary Expansion part: clinically significant changes in Leucocytes count Incidence and magnitude of clinically significant changes in Leucocytes Count (G/L) At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Red Blood Cells Count Incidence and magnitude of clinically significant changes in Red Blood Cells Count (T/L) At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Hemoglobin Incidence and magnitude of clinically significant changes in Hemoglobin (g/dL). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Hematocrit Incidence and magnitude of clinically significant changes in Hematocrit (%). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Absolute Neutrophil Count Incidence and magnitude of clinically significant changes in Absolute Neutrophil Count (G/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Absolute Eosinophil Count Incidence and magnitude of clinically significant changes in Absolute Eosinophil Count (G/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Absolute Basophil Count Incidence and magnitude of clinically significant changes in Absolute Basophil Count (G/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Absolute Lymphocytes Count Incidence and magnitude of clinically significant changes in Absolute Lymphocytes Count (G/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Absolute Monocytes Count Incidence and magnitude of clinically significant changes in Absolute Monocytes Count (G/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Platelet Count Incidence and magnitude of clinically significant changes in Platelet Count (G/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Albumin Incidence and magnitude of clinically significant changes in Albumin (g/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Bicarbonate Incidence and magnitude of clinically significant changes in Bicarbonate (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Total Bilirubin Incidence and magnitude of clinically significant changes in Total Bilirubin (µmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Calcium Incidence and magnitude of clinically significant changes in Calcium (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Urea Incidence and magnitude of clinically significant changes in Urea (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Chloride Incidence and magnitude of clinically significant changes in Chloride (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Creatinine Incidence and magnitude of clinically significant changes in Creatinine (µmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Creatinine Clearance Incidence and magnitude of clinically significant changes in Creatinine Clearance (mL/min). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Glucose Incidence and magnitude of clinically significant changes in Glucose (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Magnesium Incidence and magnitude of clinically significant changes in Magnesium (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Phosphate Incidence and magnitude of clinically significant changes in Phosphate (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Potassium Incidence and magnitude of clinically significant changes in Potassium (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Sodium Incidence and magnitude of clinically significant changes in Sodium (mmol/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Total Protein Incidence and magnitude of clinically significant changes in Total Protein (g/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Alanine aminotransferase (ALT) Incidence and magnitude of clinically significant changes in Alanine aminotransferase (ALT) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Aspartate aminotransferase (AST) Incidence and magnitude of clinically significant changes in Aspartate aminotransferase (AST) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Gamma-glutamyl transférase (GGT) Incidence and magnitude of clinically significant changes in Gamma-glutamyl transférase (GGT) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Alkaline Phosphatase (ALP) Incidence and magnitude of clinically significant changes in Alkaline Phosphatase (ALP) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Lactate dehydrogenase (LDH) Incidence and magnitude of clinically significant changes in Lactate dehydrogenase (LDH) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Amylase Incidence and magnitude of clinically significant changes in Amylase (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Lipase Incidence and magnitude of clinically significant changes in Lipase (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Total Protein Creatine Kinase (CK) Incidence and magnitude of clinically significant changes in Total Protein Creatine Kinase (CK) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Creatine kinase - cardiac muscle isoenzyme (CK-MB) Incidence and magnitude of clinically significant changes in Creatine kinase - cardiac muscle isoenzyme (CK-MB) (U/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Troponin T Incidence and magnitude of clinically significant changes in Troponin T (ng/L). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Prothrombin Time (PT) Incidence and magnitude of clinically significant changes in Prothrombin Time (PT) (Sec). At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in INR (if under VKA Therapy) Incidence and magnitude of clinically significant changes in INR (if under VKA Therapy) At the end of Cycle 1 (28 days)
Secondary Expansion part: clinically significant changes in Activated Partial Thromboplastin Time (aPTT) Incidence and magnitude of clinically significant changes in Activated Partial Thromboplastin Time (aPTT) (Sec). At the end of Cycle 1 (28 days)
See also
  Status Clinical Trial Phase
Recruiting NCT06223308 - A Study Evaluating the Safety and Efficacy of HB0028 in Subjects With Advanced Solid Tumors Phase 1/Phase 2
Not yet recruiting NCT05515185 - B7-H3 Targeting CAR-T Cells Therapy for B7-H3 Positive Solid Tumors Early Phase 1
Completed NCT05508100 - Dose Confirmation and Dose Expansion Phase 1 Study of IO-108 and IO-108 + Anti-PD-1 in Solid Tumors Phase 1
Recruiting NCT05094804 - A Study of OR2805, a Monoclonal Antibody Targeting CD163, Alone and in Combination With Anticancer Agents Phase 1/Phase 2
Completed NCT02836600 - A Study of LY3039478 in Japanese Participants With Advanced Solid Tumors Phase 1
Recruiting NCT04890613 - Study of CX-5461 in Patients With Solid Tumours and BRCA1/2, PALB2 or Homologous Recombination Deficiency (HRD) Mutation Phase 1
Recruiting NCT04390737 - Evaluate the Safety and Clinical Activity of HH2853 Phase 1/Phase 2
Recruiting NCT06007482 - A Study of ES009 in Subjects With Locally Advanced or Metastatic Solid Tumors Phase 1
Recruiting NCT05981703 - A Study Investigating BGB-26808 Alone or in Combination With Tislelizumab in Participants With Advanced Solid Tumors Phase 1
Completed NCT04108676 - Effect of Omeprazole on PK of Fluzoparib in Healthy Male Subjects Phase 1
Recruiting NCT05798611 - Study of ART0380 in Patients With Biologically Selected Solid Tumors Phase 2
Recruiting NCT05076396 - PM14 Administered Intravenously to Patients With Advanced Solid Tumors Phase 1
Recruiting NCT06008366 - A Phase 1/2 Study of 7MW3711 in Advanced Solid Tumors Phase 1/Phase 2
Recruiting NCT06054932 - Safety, Tolerability, and Immunogenicity of LK101 Alone in Participants With Incurable Solid Tumors Phase 1
Recruiting NCT04825392 - A Phase Ib Study of HX008 in Patients With Advanced Solid Tumors Phase 1
Active, not recruiting NCT04242199 - Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of INCB099280 in Participants With Advanced Solid Tumors Phase 1
Not yet recruiting NCT06365918 - Study of VG2025 Delivered Intraperitoneally in Patients With Advanced Solid Tumors With Carcinomatosis Phase 1
Recruiting NCT05443126 - A Study of EP0031 in Patients With Advanced RET-altered Malignancies Phase 1/Phase 2
Recruiting NCT05461287 - Safety, Tolerability and Pharmacokinetics Study of QLS31904 in Patients With Advanced Solid Tumors Phase 1
Recruiting NCT05569057 - A Phase I Trial of SIM1811-03 in Subjects With Advanced Solid Tumors and Cutaneous T-cell Lymphoma Phase 1