Tolerability and Safety of HF1K16 Injection in Patients With Refractory Solid Tumors

January 12, 2024 updated by: HighField Biopharmaceuticals Corporation

A Phase 1 Open-Label Dose-Escalation Study to Evaluate the Tolerability, DLT, Pharmacokinetics, and Preliminary Efficacy of HF1K16 in Patients With Refractory Solid Tumors

HF1K16 is an investigational pegylated liposome formulation of All-Trans Retinoic Acid (ATRA) for the induction of remission in patients with acute promyelocytic leukemia (APL) and for the treatment of solid tumors through targeting myeloid derived suppressor cells (MDSCs).

Study Overview

Detailed Description

Myeloid Derived Suppressor Cells (MDSCs) play important roles in constituting the immune suppressive environment promoting cancer development and progression. While previous studies had shown that all-trans retinoic acid (ATRA) could induce MDSC differentiation and maturation, the very poor solubility and fast metabolism of the drug limited its applications as an immune-modulator for cancer immunotherapy

HF1K16 is an investigational pegylated liposome formulation with great ATRA dose loading capacity and sustained drug release property. In preclinical studies, HF1K16 was shown to be able to remodel the host systemic immune homeostasis as well as modify tumor microenvironment (TME). It promotes MDSCs maturation into DCs and facilitates immune responses against cancer cells.

Study Type

Interventional

Enrollment (Estimated)

54

Phase

  • Phase 1

Contacts and Locations

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

Study Contact

Study Contact Backup

Study Locations

    • Guangdong
      • Guangzhou, Guangdong, China
        • Recruiting
        • Sun Yat-sen University Cancer Center
        • Contact:
    • Henan
      • Zhengzhou, Henan, China
        • Not yet recruiting
        • The First Affiliated Hospital of Zhengzhou University
        • Contact:
    • Hubei
      • Wuhan, Hubei, China
        • Recruiting
        • Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
        • Contact:
    • Shanghai
      • Shanghai, Shanghai, China
    • Zhejiang
      • Hangzhou, Zhejiang, China
        • Recruiting
        • Zhejiang Xiaoshan Hospital
        • Contact:
        • Contact:
      • Hangzhou, Zhejiang, China
      • Wenzhou, Zhejiang, China
        • Recruiting
        • The First Affiliated Hospital of Wenzhou Medical University
        • Contact:

Participation Criteria

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

Eligibility Criteria

Ages Eligible for Study

18 years to 75 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Inclusion Criteria:

  1. Willing and able to provide the test of informed consent in writing.
  2. Male or female, age > 18 years and < = 75 years.
  3. The subjects had to be diagnosed by histology and/or cytology with locally advanced or metastatic solid tumor. There is no effective standard of care or the patient is intolerant to the standard therapy.

    Cohort 5: The subjects must be diagnosed with glioma by histology, and the disease has relapsed or progressed after previous treatment, and there is no effective standard treatment or the subject is intolerant to standard treatment.

  4. According to the definition of RECIST 1.1, participants must have at least one measurable lesion.

    Cohort 5: at least one lesion that can be measured in two dimensions is required ( RANO criteria).

  5. Eastern group (ECOG) tumor physical state to 0 or 1. Cohort 5: According to Karnofsky physical fitness score ≥ 60.
  6. Expected lifetime > 12 weeks.
  7. Men or women of childbearing age must agree to adopt effective contraception after signing the informed consent form until 180 days after the end of the study. Premenopausal women or those within 2 years after menopause are included.

Exclusion Criteria:

  1. Patients received systemic antitumor therapy, including chemotherapy, radiotherapy, biologic therapy, endocrine therapy, or immunotherapy within 3 weeks prior to the first dose, except for the following: Nitrosoureas or mitomycin C within 6 weeks; Oral fluorouracils and small molecule drugs within 2 weeks or within 5 half-life periods of the drug (whichever is longer); Antitumour traditional Chinese medicine within 2 weeks.
  2. Adverse effects of previous anti-tumor therapy have not recovered to CTCAE 5.0 grade rating of ≤ grade 1 (except for toxicity judged by the investigator be of no safety risk, such as alopecia, grade 2 peripheral neurotoxicity, etc.)
  3. Patients received other unlisted clinical trial drugs or treatments within 28 days prior to the first dose.
  4. Taken vitamin A or any vitamin A derivatives within 7 days prior to the first dose.
  5. Past history of deep vein thrombosis or pulmonary embolism.
  6. Evidence that there is poor control of thyroid diseases, or diseases of the retina.
  7. Patients have symptomatic central nervous system (CNS) metastases, meningeal metastases, or a primary CNS tumor that is associated with progressive neurological symptoms. Except that the brain metastases are shown to be stable judged by imaging examination within 4 weeks.

    Cohort 5: The above criteria do not apply to the fifth cohort. The fifth cohort allowed inhaled or topical corticosteroids, or hormone therapy at physiological replacement doses due to adrenal insufficiency; short-term (≤7 days) corticosteroids were allowed for prophylaxis (eg, contrast media allergy) or treatment of non-autoimmune conditions ( For example, delayed-type hypersensitivity reactions caused by exposure to allergens); systemic corticosteroids (≥10 mg/day prednisone, or other equivalent corticosteroids) for 7 consecutive days within 14 days of the first dose are not allowed or Immunosuppressant therapy.

  8. Evidences of serious or uncontrolled systemic disease (for example: instability or decompensated respiratory disease, liver or kidney disease)
  9. Serious liver and kidney function damage;
  10. Has clinical significance of cardiovascular disease;
  11. Have known immune inhibitory disease or human immunodeficiency virus (HIV) infection.
  12. Patients with severe osteoporosis or with bone metastases with serum 25-hydroxyvitamin D assay values less than 50 nmol/L.
  13. Active hepatitis (Hepatitis B: HBsAg-positive and HBV-DNA ≥ 500 cps/mL or 200 IU/mLL; HCV RNA-positive).
  14. Persons with known hypersensitivity to any of the active ingredients or excipients or a history of atopic allergic reactions.
  15. The pregnancy test positive (blood beta human chorionic gonadotropin - HCG [B] test positive) or lactationWomen.
  16. Researchers believe that patients with combined disease may affect the compliance.
  17. Participants not willing to or fail to follow the procedure.
  18. Cohort 5: Brain MRI not available.
  19. Cohort 5: uncontrolled epilepsy.

Study Plan

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

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Non-Randomized
  • Interventional Model: Sequential Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Dose escalation cohort 1: HF1K16 given QOD at 45 mg/m²
The first dosing group (45 mg/m²) will include a sentinel subject receiving one dose followed by a 7-day safety evaluation interval. Three or more subjects will receive 7 doses of HF1K16 QOD at 45 mg/m2 per cycle of 21 days.
HF1K16 is a liposome suspension containing encapsulated ATRA. Drug concentration is 2mg/ml, and infusion should be completed between 60 minutes and 90 minutes
Experimental: Dose escalation cohort 2: Oral ATRA followed by HF1K16 QOD at 90 mg/m²
The second dosing group will receive oral ATRA at 45mg/m², and three days later HF1K16 QOD at 90 mg/m2 for 7 times per cycle of 21 days.
HF1K16 is a liposome suspension containing encapsulated ATRA. Drug concentration is 2mg/ml, and infusion should be completed between 60 minutes and 90 minutes
Experimental: Dose escalation cohort 3: HF1K16 QOD at 120 mg/m²
The cohort 3 will receive 7 doses of HF1K16 QOD at 120 mg/m² per cycle of 21 days.
HF1K16 is a liposome suspension containing encapsulated ATRA. Drug concentration is 2mg/ml, and infusion should be completed between 60 minutes and 90 minutes
Experimental: Dose escalation cohort 4: HF1K16 QOD at 160 mg/m²
The cohort 4 will receive 7 doses of HF1K16 QOD at 160 mg/m² per cycle of 21 days.
HF1K16 is a liposome suspension containing encapsulated ATRA. Drug concentration is 2mg/ml, and infusion should be completed between 60 minutes and 90 minutes
Experimental: Dose escalation cohort 5: HF1K16 QOD at 120mg or180 mg
The cohort 5 will receive 7 doses of HF1K16 QOD at 120 mg or 180mg per cycle of 21 days.
HF1K16 is a liposome suspension containing encapsulated ATRA. Drug concentration is 2mg/ml, and infusion should be completed between 60 minutes and 90 minutes

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Incidence of Adverse Events
Time Frame: 30 days after administration
Defined by the Common Terminology Criteria for Adverse Events version 5.0 (CTCAE V5.0)
30 days after administration
Incidence of dose-limiting toxicities(DLT)
Time Frame: 21 days after administration
Observe the dose limiting toxicity, and Incidence of dose-limiting toxicities(DLT) will be assessed
21 days after administration
Respiration rate of Vital Signs by stethoscope
Time Frame: 30 days after administration
Changes from baseline for respiration rate in breaths per minute of Vital Signs
30 days after administration
Red blood cell count in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for Red blood cell count in whole blood in10^9 /L
30 days after administration
Ventricular rate of ECG
Time Frame: 30 days after administration
Changes from baseline for ventricular rate in beats per minute
30 days after administration
Respiration rate in mg μl/h·g of ECG
Time Frame: 30 days after administration
Changes from baseline for respiration rate in mg μl/h·g
30 days after administration
Heart rate in beats per minute in beats per minute of ECG
Time Frame: 30 days after administration
Changes from baseline for heart rate in beats per minute
30 days after administration
Blood pressure by sphygmomanometer
Time Frame: 30 days after administration
Changes from baseline for blood pressure in mmHg, both systolic and diastolic pressures will be assessed.
30 days after administration
Body temperature by thermometer
Time Frame: 30 days after administration
Changes from baseline for body temperature in Celsius degree
30 days after administration
White blood cell count in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for white blood cell count in whole blood in 10^9 /L
30 days after administration
Neutrophil count in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for neutrophil count in whole blood in 10^9 /L
30 days after administration
Hemoglobin concentration in g/dL in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for hemoglobin concentration in g/dL in whole blood
30 days after administration
Prothrombin time in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for Prothrombin time in s
30 days after administration
International standardized ratio in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for international standardized ratio
30 days after administration
International sensitivity index in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for international sensitivity index
30 days after administration
Activated partial thromboplastin time in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for activated partial thromboplastin time in s
30 days after administration
Total bilirubin concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for total bilirubin concentration in μmol/L
30 days after administration
ALT concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for alanine aminotransferase(ALT) concentration in U/L
30 days after administration
AST concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for aspartate aminotransferase(AST) concentration in U/L
30 days after administration
Total protein concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for total protein concentration in g/L
30 days after administration
Urea concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for urea concentration in mmol/L
30 days after administration
Creatinine concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for creatinine concentration in μmol/L
30 days after administration
Total cholesterol concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for total cholesterol concentration in mmol/L
30 days after administration
Triglycerides concentration in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for triglycerides concentration in mmol/L
30 days after administration
HDL-C in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for high density lipoprotein cholesterol (HDL-C) in mmol/L
30 days after administration
LDL-C in whole blood sample
Time Frame: 30 days after administration
Changes from baseline for low density lipoprotein cholesterol (LDL-C) in mmol/L
30 days after administration
PR interval by ECG
Time Frame: 30 days after administration
Changes from baseline for PR interval in ms of ECG
30 days after administration
QRS by ECG
Time Frame: 30 days after administration
Changes from baseline for QRS in ms of ECG
30 days after administration
QT by ECG
Time Frame: 30 days after administration
Changes from baseline for QT in ms of ECG
30 days after administration
QTc by ECG
Time Frame: 30 days after administration
Changes from baseline for QTc in ms of ECG
30 days after administration
Cohort 5: Determination of overall response rate (ORR) according to RANO criteria
Time Frame: From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
ORR is defined as the proportion of participants with complete response or partial response (CR+PR)
From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
Cohort 5: Duration of response (DOR)
Time Frame: From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
DOR, defined as the time between the start of the subject's first assessment of CR or PR and the first assessment of PD or death from any cause, according to RANO criteria by the investigator
From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
Cohort 5: disease control rate (DCR)
Time Frame: From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
DCR , defined as the proportion of subjects with a best overall response of CR, PR, or SD in the study as assessed by the investigator according to RANO criteria
From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
Cohort 5: progression-free survival (PFS).
Time Frame: From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks
PFS, defined as the time between the subject's first dose and the onset of (any aspect of) tumor progression or death from any cause
From date of first dose until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 36 weeks

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
HF1K16 pharmacokinetic parameters with Cmax
Time Frame: Up to 48 hours postdose
Maximum plasma concentration (Cmax) after administration of HF1K16
Up to 48 hours postdose
the overall response rate(ORR) of HF1K16
Time Frame: Once every six weeks in the first year, then once every 12 weeks afterwards through study completion, an average of 1 year
ORR is defined as the proportion of participants with complete response or partial response (CR+PR)
Once every six weeks in the first year, then once every 12 weeks afterwards through study completion, an average of 1 year
Peripheral blood mononuclear cells by whole blood sample
Time Frame: Before injection on Day1,7,13, 21 in each cycle(each cycle is 21 days)
Assessing peripheral blood mononuclear cells after dose in cells/mL
Before injection on Day1,7,13, 21 in each cycle(each cycle is 21 days)
AUC48h by plasma concentration of whole blood sample
Time Frame: Up to 48 hours postdose
Area under plasma concentration -time curve from 0 time ot 48 h(AUC0-48) after dose
Up to 48 hours postdose
Tmax by plasma concentration of whole blood sample
Time Frame: Up to 48hours postdose
Peak time (Tmax) after dose
Up to 48hours postdose
T1/2 by plasma concentration of whole blood sample
Time Frame: Up to 48hours postdose
Elimination half-life (T1/2) after dose
Up to 48hours postdose
CL by plasma concentration of whole blood sample
Time Frame: Up to 48 hours postdose
Clearance (CL) after dose
Up to 48 hours postdose
Vd by plasma concentration of whole blood sample
Time Frame: Up to 48 hours postdose
Volume of distribution(Vd) after dose
Up to 48 hours postdose
AUClast by plasma concentration of whole blood sample
Time Frame: Up to 48 hours postdose
Ratios of geometric means of AUClast (Area under the plasma concentration-time curve from zero to time of last quantifiable concentration) after dose
Up to 48 hours postdose
Cohort 5: To assess the changes in MDSC after HF1K16 treatment, including changes in MDSC number
Time Frame: Before injection on Day1 in each cycle(each cycle is 21 days)
Assessing peripheral blood mononuclear cells after dose in cells/mL
Before injection on Day1 in each cycle(each cycle is 21 days)
Cohort 5: Incidence of Adverse Events
Time Frame: after administration
Defined by the Common Terminology Criteria for Adverse Events version 5.0 (CTCAE V5.0)
after administration
Cohort 5: To assess the changes in MDSC after HF1K16 treatment, including changes in MDSC phenotype
Time Frame: Before injection on Day1 in each cycle(each cycle is 21 days)
Assessment of MDSC phenotype in peripheral blood after drug administration
Before injection on Day1 in each cycle(each cycle is 21 days)

Collaborators and Investigators

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

Investigators

  • Principal Investigator: Jinsong Wu, MD, Huashan Hospital Shanghai

Study record dates

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

Study Major Dates

Study Start (Actual)

February 16, 2022

Primary Completion (Estimated)

April 1, 2025

Study Completion (Estimated)

November 1, 2025

Study Registration Dates

First Submitted

May 4, 2022

First Submitted That Met QC Criteria

May 18, 2022

First Posted (Actual)

May 24, 2022

Study Record Updates

Last Update Posted (Actual)

January 16, 2024

Last Update Submitted That Met QC Criteria

January 12, 2024

Last Verified

January 1, 2024

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

Yes

Studies a U.S. FDA-regulated device product

No

product manufactured in and exported from the U.S.

No

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

Clinical Trials on Solid Tumor, Adult

Clinical Trials on HF1K16 /Arm 45 mg/m²

3
Subscribe