STRENGTHS AND LIMITATIONS OF THIS STUDY
-
In the context of difficult-to-treat diseases that lack effective standard treatment, initially explore reliable therapies.
-
Quantitative and qualitative data are collected at fixed points in time for subsequent analysis.
-
The early clinical data of therapeutic drugs suggest that their safety risks are predictable, acceptable and controllable.
-
The main limitation of this study is the single-arm experiment, which is compared with external historical data, weakening the strength of the argument.
-
In the absence of randomised parallel controls, there are many uncertainties when using the results of single-arm studies as the basis for benefit–risk assessment.
Introduction
Primary liver cancer is a malignant tumour that critically threatens human health, and global rates of its incidence and fatality, respectively, rank sixth and third.1 Hepatocellular carcinoma (HCC) is the most common histological type of liver cancer and accounts for approximately 90% of cases worldwide.2 The vast majority of patients with HCC have latent onset and are in the middle and late stages of initial diagnosis.3 Only approximately 10% of patients are considered for radical resection,4 and more than 50% of postoperative patients have short-term recurrence.5 6 Patients with HCC have a poor prognosis, and untreated patients have a short survival, with median overall survival of less than 10 months.7 8 Therefore, prolonging the survival time of patients with HCC and improving the quality of life of patients are urgent problems to be solved.
In 2007, the tyrosine kinase inhibitor (TKI) sorafenib was approved as the only available systemic treatment for advanced HCC.9 10 In recent years, new drugs such as regorafenib have proven to be as effective and have been successively approved for advanced HCC treatment.11 12 With the discovery of immune checkpoint inhibitors (ICIs), significant breakthroughs have been made in HCC treatment.13 14 The safety and efficacy of combination therapy with programmed cell death 1 (PD-1)/PD-1 ligand (PD-L1) inhibitors and molecularly targeted drugs have been demonstrated.15–17 This paradigm shift in treatment suggests that PD-1/PD-L1 combined with antiangiogenic drugs may be the key to effective treatment of advanced HCC.
In 2019, a global phase 3 trial named IMbrave150 showed that patients with advanced HCC with the first-line treatment of atezolizumab combined with bevacizumab had survival benefit of approximately 6 months longer than patients with sorafenib monotherapy.18 Subsequently, Orient-32, a multicentre, randomised phase 3 trial in China, demonstrated significant overall survival (OS) and progression-free survival (PFS) benefits with acceptable safety for the first-line treatment of unresectable or advanced HCC with sintilimab combined with IBI305.19 In addition, a phase Ib single-arm study, KEYNOTE524 showed that lenvatinib and pembrolizumab, as first-line regimens, demonstrated outstanding antitumor activity in the treatment of advanced unresectable HCC, with disease control rate (DCR) >85%, median PFS of 9.3 months and median OS of 22.0 months.20 However, these trials were conducted in patients with advanced HCC without previously received systemic therapy, meaning that patients who had received sorafenib or other first-line therapies were excluded. The American Society of Clinical Oncology (ASCO) guidelines recommend that atezolizumab plus bevacizumab can be considered as a second-line therapy in patients who have failed first-line sorafenib or lenvatinib therapy.21 A retrospective study reported that patients treated with atezolizumab plus bevacizumab after first-line therapy had a survival time of approximately 16 months, which is approximately 5–8 months longer than patients treated with the approved drugs.22 However, this treatment regimen has not reached a formal consensus nor has it been included in any HCC diagnosis and treatment guidelines, and its application still needs to be supported by more reliable research data.
Traditional radiotherapy (RT) is limited in the treatment of HCC due to its simple positioning, large radiation area in normal liver tissue and easy to induce radioactive liver disease.23 24 However, with the rapid development of precision RT techniques such as three-dimensional conformal RT and stereotactic body RT (SBRT), RT has shown good local control rates and safety in HCC treatment. The 1-year, 2-year and 3-year local control rates of SBRT for the treatment of liver cancer are 93%, 89% and 86%, respectively, and the incidence of severe toxicity is usually less than 10%, making RT one of the main treatment methods for liver cancer.25 26 In contrast, studies have shown that RT has the distant effect on tumours, and immunotherapy can mediate and enhance this effect, indicating that RT and immunotherapy have a synergistic effect, and the combination of the two may be a promising treatment for advanced liver cancer.27 28 Multiple studies have shown that RT combined with a PD-1/PD-L1 monoclonal antibody has high efficacy and safety and is a promising treatment strategy for HCC.29–31 The results of a real-world clinical study demonstrate that the combination of SBRT based on targeted therapy and immunotherapy can serve as an efficacious conversion therapy for patients with HCC with extrahepatic metastasis, thereby potentially extending survival.32
Although the results of similar studies seem encouraging, more prospective trials are needed to confirm the exact efficacy of RT in combination with targeted therapy and immunotherapy.
In summary, we designed and conducted a single-arm, single-centre, open-label phase 2 clinical study to evaluate the efficacy and safety of SBRT in combination with sintilimab and IBI305 as a treatment option for patients with advanced HCC who have received at least one previous immunotherapy.
Methods and analysis
Study design
This single-arm, open-label phase 2 trial will be conducted at the Union Hospital Affiliated with Tongji Medical College of Huazhong University of Science and Technology in Wuhan, China. The study was authorised by the Medical Ethics Committee, and all patients will provide written informed consent before enrolment. The trial was registered in the Chinese Clinical Trials Registry under the number ChiCTR2200056068 (online supplemental files 1–3).
Supplemental material
Supplemental material
Supplemental material
Recruitment began in February 2022 and is expected to continue until July 2023. Eligible patients with advanced HCC will be treated with SBRT, sintilimab and IBI305. We will use the objective response rate (ORR) as the primary endpoint and ORR of lesions without RT, DCR, PFS, OS and adverse events (AEs) as secondary endpoints.
Participant selection
In this study, patients with HCC will be diagnosed according to histopathology or clinical criteria of the European Association for the Study of the Liver guidelines33 or diagnostic criteria of the Chinese Diagnosis and Treatment Standards for Primary Liver Cancer (2019 Edition). Participants will be selected based on the following inclusion and exclusion criteria.
Inclusion criteria
-
Male or female ≥18 years old;
-
Eastern Cooperative Oncology Group Performance Status (ECOG PS) Score from 0 to 1;
-
According to the Chinese Code of Diagnosis and Treatment for Primary Liver Cancer (2019 version), it is defined as inoperable advanced HCC;
-
Have received at least first-line systemic antitumour therapy, including PD-1 inhibitor monotherapy or a combination regimen for HCC;
-
Eligible to receive radiation therapy;
-
To have at least one measurable lesion according to RECIST1.1 and at least one measurable lesion without RT;
-
Child-Pugh grade A;
-
Expected survival time >3 months;
-
Have full organ and bone marrow function, as follows:
-
Adequate blood function: absolute neutrophil count ≥1.5×109 /L; platelet count ≥60×109 /L; haemoglobin content ≥90 g/L.
-
Adequate liver function: serum total bilirubin ≤3× ULN (upper limit of normal); alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate transferase and ALP ≤5× ULN; serum albumin ≥30 g/L.
-
Adequate renal function: serum creatinine ≤1.5× ULN or clearance of creatinine ≥50 mL/min (Cockcroft-Gault formula); routine urine results show urine protein <2+; for patients with urine protein ≥2+ in routine urine tests at baseline, 24-hour urine collection and 24-hour protein quantity <1 g should be performed.
-
Adequate coagulation function: international normalised ratio and activated partial thromboplastin time ≤1.5 times ULN.
-
Normal thyroid function, with total triiodothyronine (T3), free T3 and free thyroxine levels within the normal range (this can be controlled with thyroid replacement therapy).
-
-
Male and premenopausal women: consent to use a medically approved contraceptive during the study treatment period and for 6 months after the end of the study treatment period. The female patient was not lactating and had a negative blood or urine pregnancy test within 7 days prior to enrolment.
Exclusion criteria
-
History of hepatic encephalopathy or liver transplantation;
-
History of autoimmune diseases and autoimmune defects;
-
Presence of clinical symptoms requiring drainage of pleural fluid, ascites and pericardial effusion;
-
Infection of acute or chronic active hepatitis B or C; hepatitis B virus DNA >2000 IU/mL or 104 copies/mL, hepatitis C virus (HCV) RNA>103 copies/mL, hepatitis B surface antigen and anti-HCV antibodies both positive;
-
Central nervous system metastasis;
-
Prior RT for liver disease;
-
Poor blood pressure control despite medication: systolic >140 mm Hg or diastolic >90 mm Hg;
-
Bleeding events of oesophageal or fundus varices caused by portal hypertension that occurred within the past 6 months. Severe (G3) varicose veins observed on endoscopy within 3 months of the initial dosing; Patients with evidence of portal hypertension (including splenomegaly on imaging) and at high risk of bleeding as assessed by the investigator;
-
Any life-threatening bleeding event requiring transfusion therapy, surgery or local therapy, or ongoing medication occurred in the 3 months prior to recruitment;
-
Arterial or venous thromboembolism events in the 6 months prior to recruitment, including myocardial infarction, unstable angina pectoris, cerebrovascular accident or transient ischaemic attack, pulmonary embolism, deep vein thrombosis or any other serious thromboembolism; Implantable intravenous infusion port or catheter-derived thrombosis, or superficial venous thrombosis, except in cases where the thrombus has stabilised after conventional anticoagulant therapy; Prophylactic use of low dose low molecular weight heparin (eg, enoxaparin 40 mg/day) is permitted;
-
Severe bleeding tendency or clotting dysfunction, or being treated with thrombolysis;
-
Previous or current pulmonary diseases such as pulmonary fibrosis, interstitial pneumonia, pneumoconiosis, drug-related pneumonia and severe impairment of lung function;
-
Patients who have active tuberculosis (TB) are receiving anti-TB therapy or have received anti-TB therapy within 1 year prior to first dosing;
-
Patients with HIV infection (HIV 1/2 antibody positive) or known syphilis infection;
-
Patients with severe infection who are active or poorly controlled. Severe infection within 4 weeks prior to initial dosing, including but not limited to hospitalisation due to complications of infection, bacteremia or severe pneumonia;
-
Immunosuppressive drugs have been used within 4 weeks prior to initial dosing, excluding nasal, inhaled or other routes of topical corticosteroids and physiological doses of systemic corticosteroids (not exceeding 10 mg/day of prednisone or equivalent doses of other corticosteroids), allowing temporary use of corticosteroids to treat symptoms of breathing difficulties in asthma, chronic obstructive pulmonary disease, etc;
-
Received Chinese medicines with antitumour indications or immunomodulatory effects (including thymosin, interferon, interleukin, except for topical use to control pleural fluid or ascites) within 2 weeks prior to initial dosing;
-
Other malignancies were diagnosed within 5 years prior to initial dosing, excluding basal cell carcinoma of the skin, squamous cell carcinoma of the skin and/or carcinoma in situ that had been radically resected;
-
Patients who are known to be allergic to any of the components of sintilimab, bevacizumab biosimilar (IBI305) formulations or have had prior severe allergic reactions to other monoclonal antibodies or vascular endothelial growth factor A (VEGFA) inhibitors;
-
Received treatment in other clinical trials within 4 weeks prior to initial dosing;
-
Pregnant or nursing female patients;
-
Other acute or chronic medical conditions, psychiatric disorders or abnormalities in laboratory test values that may increase the risk associated with study participation or study drug administration, or interfere with the interpretation of study results, and the investigator determines that the patient is ineligible to participate in the study.
Interventional methods
Immunotherapy
Sintilimab will be administered intravenously at a dose of 200 mg every 21 days. The maximum duration of treatment is 24 months. The first dose of sintilimab will be administered within 3 days of treatment initiation and continue until progressive disease (PD) or intolerable toxicity is observed. If AEs, abnormalities in laboratory tests or other comorbidities occur during treatment, sintilimab will be delayed or suspended; however, dose reduction is not permitted. Patients will be withdrawn from the study if treatment is interrupted for more than 12 weeks (continuous or intermittent). If treatment-related adverse reactions do not return to grades 0–1 or to baseline within 6 weeks of the last dose of sintilimab, it should be permanently discontinued.
Targeted therapy
IBI305 will be administered intravenously at a dose of 15 mg/kg every 21 days, each time after sintilimab administration at least 5 min apart, and continued until the patient develops PD or intolerable toxicity. Dosing adjustments for IBI305 are not allowed during treatment. The dose of IBI305 will be converted from the subject’s weight at baseline (prior to day 1 administration) and remain constant throughout the study unless the patient’s weight changes by more than 10% from baseline.
Radiotherapy
SBRT will start within 7 days of participation in the trial. The RT programme was completed with 4D CT to evaluate tumour movement. General tumour volume (GTV) (including primary and metastatic tumours) will be defined by contrast-enhanced CT or MRI if dose–volume limits for organs at risk can be met.34 And increasing GTV by 5–10 mm produces clinical tumour volume (CTV). The planning target volume will be constructed by adding 5–10 mm of the CTV in all directions. Suitable lesions were selected and short-range RT was performed with 6 MV X-ray linear accelerator at a dose of 30–40 Gy/5–8 times, 5 times a week until the end of RT. The specific RT regimen will be determined by the investigator.
Assessment
Tumour response assessment
Baseline tumour imaging scans must be performed within 28 days prior to initial dosing. Tumour radiological evaluations will be performed at 6-week intervals (±7 days), beginning with the initial administration. After 48 weeks of treatment, radiological tumour assessments will be performed at 12-week intervals (±7 days). The radiological methods used to evaluate the tumour at baseline must be consistent with those used for each subsequent follow-up evaluation, including but not limited to chest CT and abdominal MRI. Examination of other affected sites is permitted as indicated by the signs and symptoms of each patient.
Based on RECIST version 1.1,35 patients in the trial group who showed evidence of radiological PD at the initial efficacy evaluation will be allowed to continue treatment if the investigator considers that the patients would continue to gain clinical benefit from the study protocol and are re-evaluated for radiological confirmation every 4 weeks (±7 days). If PD is confirmed, the patients should discontinue the study protocol; if no progress is confirmed, the protocol therapy should continue to be studied and evaluated according to the scheduled time point of imaging examination specified in the protocol until PD is confirmed on imaging. For patients first assessed by the investigator as having stable disease (SD), partial response (PR) or complete response (CR), treatment and tumour evaluation will continue until treatment is discontinued.
If the patient becomes clinically unstable during the study, unscheduled radiological evaluation may be performed at any time. Clinical disease instability is defined as follows: (1) presence of clinically significant signs and symptoms suggestive of disease progression (including deterioration of laboratory test values); (2) decreased ECOG PS Score; (3) rapid disease progression and (4) tumour progression at important anatomical sites requiring other urgent medical intervention (eg, spinal cord compression).
For patients who discontinue treatment for reasons other than radiological disease progression, radiological evaluation should be performed at the end of treatment and subsequently continued at planned time points specified in the protocol until any of the following events occur: initiation of new antitumour therapy, objective disease progression, withdrawal of informed consent, loss to follow-up or death.
Safety assessment
All AEs will be evaluated according to the National Cancer Institute (NCI) General Terminology for Adverse Events (CTCAE) version 5.0.36 AEs are defined as any adverse or undesired medical events occurring after the subjects of the clinical study signed the informed consent form, regardless of whether there was a causal relationship with the study drug. AEs include but are not limited to the following:
-
An increase in pre-existing (pretrial) medical conditions/diseases (including an increase in symptoms, signs and abnormalities in laboratory tests);
-
Any newly occurring adverse medical conditions (including symptoms, signs and newly diagnosed diseases);
-
Abnormal laboratory results of clinical significance.
Serious AEs (SAEs) are those that meet at least one of the following criteria:
-
Cause death, other than death caused by progression of disease for study indications;
-
Life-threatening (‘life threatening’ in the definition means that the subject is at risk of death when this AE occurs and does not include AE that would have caused death if the event had been aggravated);
-
Hospitalisation or prolonged hospitalisation is required;
-
Permanent or serious disability/disability;
-
Congenital abnormalities/birth defects;
-
Other medically significant events are defined as events that endanger the patients or require medical intervention to prevent any of the above events.
Any AEs will be fully documented, including the diagnosis (if there is no diagnosis, the symptoms and signs, including abnormal lab tests), start and end dates and times (if applicable), CTCAE severity rating and change (grade 3 or above events), whether there were SAEs, measures taken with study drugs, outcomes of treatments and events given for AE and the relationship between AEs and study drugs.
AEs should be followed-up until recovery to baseline, grade 0 to 1 or if the investigator believes for reasonable reasons (such as no recovery or improvement), further follow-up is not necessary.
Statistical analysis
Sample size
The sample size was calculated using Simon’s two-stage design. Previous studies have shown that the ORR of second-line TKI or immunotherapy for liver cancer is between 10.6% and 24.1%,37 and it is estimated that the ORR in this study can reach 25%. We plan to enrol 21 subjects, and the 95% CI can be maintained within the range of 8.7%–49.1% (figure 1).
Statistical analysis methods
All statistical analyses will be performed using SAS V.9.2 (later version) statistical analysis software. All statistical tests will be performed using a one-sided validity hypothesis test with p<0.05. The 95% CIs and p values will be provided for comparison between groups. Unless otherwise stated, measurement data will be statistically described as mean±SD or median (minimum, maximum). The frequency (percentage) is used for the statistical description of the counting data. Kaplan-Meier curves will be plotted to compare OS and PFS between the two groups using the log-rank test. The duration of the overall response (DOR) and ORR will be calculated based on binomial distributions using the two-sample Cochran-Mantel-Haenszel method.
Outcome definitions
Efficacy analysis
-
The ORR is defined as the proportion of subjects with CR and PR to the total number of participants;
-
The DCR is defined as the proportion of subjects with CR, PR and SD;
-
PFS, defined as the time from enrolment to the first progression of a radiological disease or death, whichever occurs first;
-
OS, defined as the time from enrolment to death due to any cause;
-
The DOR is the time between the first evaluation of a tumour as CR or PR and the first evaluation as PD or death from any cause.
ORR, PFS, DCR and DOR will be evaluated by the investigator according to RECIST v1.1 criteria.
Safety analysis
In this study and during follow-up, the severity of AEs will be graded according to NCI CTCAE (version 5.0). The safety evaluation indicators include the following:
-
Incidence and severity of all AEs; treatment-phase AEs, SAEs, immune-related AEs and their associations with study drugs;
-
Number and proportion of subjects who discontinue treatment owing to the above AEs;
-
Changes in vital signs, physical examination and laboratory results before, during and after treatment.
Patient and public involvement
Patients or the public were not involved in the design, implementation, reporting or dissemination of our studies.
Discussion
In recent years, the advent of ICIs has brought new hope to the treatment of HCC.14 15 ICIs, antiangiogenic antibodies and other targeted drugs have resulted in breakthroughs in treating various tumours.16 17 38 39 Based on several clinical studies,18–20 40 domestic and foreign authoritative guidelines recommend PD-1/PD-L1 combined with TKI/bevacizumab as a first-line treatment for HCC. A real-world study of 60 patients showed that PD-1 inhibitors combined with TKI for unresectable HCC had an ORR of 36.7% (95% CI 24.9% to 48.5%) and DCR of 81.7% (95% CI 73.2% to 93.8%).41
Although ICIs combined with targeted therapy have made remarkable achievements in the first-line treatment of HCC, the effect of immunotherapy on HCC is limited, with an effective rate of less than 20% for monotherapy and 30%–36% for combination therapy.20 42 43 In addition, some patients develop resistance to ICI therapy. With the popularity of first-line ICIs combined with TKI therapy, the follow-up treatment of patients with HCC has faced new challenges. Currently, the second-line optional regorafenib, PD-1 inhibitors or Ramolumab are all targeted at patients who have developed first-line sorafenib treatment.21 44–46 At present, there are no clear guidelines and valid data support for ICIs combined with TKI as the best second-line drug regimen for patients with advanced HCC.
Through direct and indirect killing effects, RT can gradually induce apoptosis and necrosis in tumour cells. In theory, the continuous release of tumour antigens can be achieved throughout the entire process of RT and within a few months after RT, which is one of the most effective ways to induce active immunity in the body and is highly likely to enhance the immunogenicity of liver cancer and improve the efficacy of immunotherapy.47 Preclinical studies have shown that the combination of anti-PD-L1 and radiation can enhance the antitumour effects of radiation in mouse HCC.28 48 Chiang et al reported nivolumab immunotherapy after SBRT achieved 100% ORR in five patients with large, unresectable HCC.49 In a retrospective study, Smith et al evaluated the safety and efficacy of nivolumab after RT or in combination with unresectable hepatocellular an ORR of 14.8%.50 Another study on second-line and third-line treatments for liver cancer showed that 65.7% ORR (5.9% CR, 58.8% PR) and 88.2% DCR were achieved in 17 patients treated with toripalimab during or after RT.51 Multiple studies have suggested that RT combined with ICIs may be effective in the treatment of advanced recurrent HCC.
However, few prospective studies have been conducted on PD-1/PD-L1 combined with TKI/bevacizumab in patients with HCC who are insensitive to first-line immunotherapy. To explore the best second-line treatment plan for patients with HCC who have progressed after first-line immunotherapy, this study aims to evaluate the efficacy and safety of SBRT combined with sintilimab and IBI305 treatment, which is believed to bring more hope for the follow-up treatment of patients with advanced HCC.
Limitation
The main limitation of this study was the single-arm trial. Due to the lack of a standard effective treatment, it is not possible to set up a randomised control group but to use historical external data controls, which increases the uncertainty in assessing the benefits of treatment. However, phase 2 trials are more exploratory than confirmatory, and regression of malignant tumours by itself is almost impossible, the tumour response can be assumed to be almost entirely the effect of the drug. This study used the Simon two-stage design, which aims to test whether the treatment had an advantage at an early stage of the trial. If the expected effect is achieved initially, further study will be conducted; otherwise, the study of this experimental group will be terminated to avoid more subjects with ineffective treatment and waste of resources.
Ethics statements
Patient consent for publication
Acknowledgments
We acknowledge and express gratitude for the active collaboration and support provided by the physicians from the Department of Oncology, the Department of Radiology, the Department of Pathology and the Department of Gastroenterology of Wuhan Union Hospital in the development of this study protocol.
This post was originally published on https://bmjopen.bmj.com