Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Sep 1.
Published in final edited form as: Cancer. 2024 Feb 15;130(17):2918–2927. doi: 10.1002/cncr.35243

SWOG 1609 Cohort 48: Anti–CTLA-4 and Anti–PD-1 for Advanced Gallbladder Cancer

Sandip P Patel a,*, Elizabeth Guadarrama b, Young Kwang Chae c,*, Michael J Dennis a, Benjamin C Powers d, Chih-Yi Liao e, William A Ferri Jr f, Thomas J George g, Elad Sharon h, Christopher W Ryan i, Megan Othus j, Gabby Lopez j, Charles D Blanke k, Razelle Kurzrock l,*
PMCID: PMC11309904  NIHMSID: NIHMS1979911  PMID: 38358334

Abstract

Introduction:

Most patients with advanced gallbladder cancer are treated with multi-agent chemotherapy. Immune checkpoint inhibitors offer the possibility of a durable response with less toxicity. This prospective, multicenter, open-label study was designed to evaluate the anticancer activity of nivolumab plus ipilimumab in patients with advanced gallbladder cancer.

Methods:

Nineteen patients with advanced gallbladder cancer refractory to ≥ 1 prior therapy received nivolumab 240 mg intravenously every 2 weeks and ipilimumab 1 mg/kg intravenously every 6 weeks until disease progression or unacceptable toxicity. The primary endpoint was confirmed radiographic overall response rate (ORR) [complete response (CR) + partial response (PR) confirmed on subsequent scan); secondary endpoints included unconfirmed overall response, clinical benefit rate (confirmed and unconfirmed responses + stable disease >6 months), progression-free survival (PFS), overall survival, and toxicity.

Results:

The confirmed ORR was 16% [CR, n=1 (5%); PR, n=2 (11%)]; all were microsatellite stable, and the confirmed CR had undetectable PD-L1 by immunohistochemistry. The unconfirmed ORR and clinical benefit rate were both 32%. The median duration of response was 14.8 months (range, 4–35.1+ months). The 6-month PFS was 26% (95% CI, 12–55%). The median overall survival was 7.0 months (95% CI, 3.9–19.1 months). The most common toxicities were fatigue (32%), anemia (26%), and anorexia (26%). Aspartate aminotransferase elevation was the most common grade 3/4 toxicity (11%). There was one possibly related death (sepsis with attendant hepatic failure).

Conclusions:

Ipilimumab plus nivolumab was well tolerated and showed modest efficacy with durable responses in previously treated patients with advanced gallbladder cancer.

Keywords: Immune checkpoint inhibitor, biliary tract cancer, nivolumab, ipilimumab

Précis:

Ipilimumab plus nivolumab demonstrated modest efficacy with durable responses and manageable toxicity in previously treated patients with advanced gallbladder cancer.

Plain Language Summary:

This prospective study assessed the efficacy and safety of nivolumab plus ipilimumab in 19 patients with advanced gallbladder cancer refractory to prior therapy. The combination demonstrated modest efficacy with a 16% confirmed overall response rate, durable responses, and manageable toxicities, suggesting potential benefits for this challenging patient population.

Introduction

Gallbladder cancer is a rare but aggressive biliary tract cancer, accounting for 0.6% of all cancer diagnoses globally1. Symptoms and signs of gallbladder cancer are not specific, and often appear late in the clinical course of disease2. For this reason, patients are frequently diagnosed at an advanced stage, when the prognosis is poor and treatment options are limited.

Until recently, the standard first-line treatment for advanced biliary tract cancer was the combination of gemcitabine and cisplatin, which demonstrated improved overall survival (OS) compared to gemcitabine alone in the ABC-02 trial (NCT00262769)3. This regimen was only recently supplanted by a new standard based on the results of two phase III trials, TOPAZ-1 (NCT03875235) and KEYNOTE-966 (NCT04003636)4,5. Both trials combined immune checkpoint inhibition with gemcitabine and cisplatin and showed an improvement in median OS compared to gemcitabine and cisplatin in the first-line. While this was considered a success, the reality is that most patients will experience progression at some point in their treatment course, requiring consideration of a subsequent line of therapy.

There has been growing interest in the identification of molecularly-targeted therapies for biliary tract cancer, particularly in the relapsed/refractory and advanced settings. This is perhaps most notable with the drug approvals for both fibroblast growth factor receptor (FGFR) and isocitrate dehydrogenase-1 (IDH1) inhibitors for cholangiocarcinoma. Unfortunately, gallbladder cancer was not included in the trials that led to these approvals, leaving question as to whether the positive findings would be applicable to all biliary tract cancers6,7. Alterations in these genes are also extremely rare in gallbladder cancer (<5%), as are the required molecular alterations for other drugs with tissue-agnostic approvals810. There is hope that human epidermal growth factor receptor 2 (HER2)-directed therapies will soon be available for the 16% of patients with gallbladder cancer and HER2 amplification, noting two recent phase II trials that showed efficacy9,11,12. However, the majority of patients with advanced gallbladder cancer will continue to be treated with combination chemotherapy.

ABC-06 (NCT01926236) established FOLFOX (folinic acid, fluorouracil, and oxaliplatin) as the standard of care subsequent-line therapy for relapsed/refractory biliary tract cancer13. There was no prior standard of care, and the control arm was active symptom control. While FOLFOX was able to demonstrate efficacy, improving the median OS from 5.3 months to 6.2 months, the objective response rate (ORR) was only 5%. There was also substantial toxicity. Smaller scale studies with alternative regimens had similar outcomes with response rates ranging 0–15%1416.

A few trials now exist examining the role of immune checkpoint inhibitors (ICIs) in relapsed/refractory biliary tract cancer. This includes pembrolizumab alone or in combination with lenvatinib, nivolumab monotherapy, and durvalumab alone or in combination with tremelimumab1720. These studies yielded ORRs of 4.8–13% with median overall survivals of 5.7–14.2 months. Pembrolizumab also showed efficacy in microsatellite instability-high (MSI-High) advanced biliary tract cancer, where the ORR was 40.9% and the median overall survival was 24.3 months21. This later study highlights the importance of MSI testing in biliary tract cancer, and also explains at least part of the variability observed in the other trials, where MSI status was not always reported1720. Another key observation was that some patients had a duration of response (DOR) extending beyond 2 years, an achievement that had not been seen with other therapies [median progression-free survival (PFS) in ABC-06 (which used FOLFOX) was 4.0 months; 95% confidence interval (CI), 3.2–5.0 months; DOR was not reported]13.

The combination of nivolumab and ipilimumab has also been studied in the Australian phase II CA209–538 trial designed for patients with rare cancers, including a cohort of advanced biliary tract cancers22. The ORR was modest in the biliary tract cohort (23%), but higher in the subgroup of patients with advanced gallbladder cancer. The majority of patients in this study had 1 prior line of systemic treatment (range 0–2). Biomarker assessment was not broadly conducted, but all responding patients were microsatellite stable.

We now present the data from the gallbladder cancer cohort of SWOG 1609 dual anti-CTLA-4 & anti-PD-1 blockade in rare tumors (DART; NCT02834013), a basket immunotherapy trial studying ipilimumab plus nivolumab across multiple cohorts of rare tumors. This study, while similar to CA209–538, differed in regards to patient population. In particular, SWOG 1609 required all patients to be treated with at least one prior line of therapy. This resulted in a more refractory patient population with an overall poorer performance status. The SWOG 1609 gallbladder cohort also required patients to have a diagnosis of gallbladder cancer (as opposed to CA209–538 where 67% of the subjects had a biliary tract cancer outside the gallbladder). These differences should be considered when reviewing the data presented herein.

Materials and Methods

Study design and eligibility criteria

S1609 DART is an open-label, multi-institutional, single-arm phase II basket study designed to evaluate the response of rare tumors to the combination of ipilimumab and nivolumab. Rare cancers were defined as having an incidence of less than 6 in 100,000 per year23. Patients with gallbladder cancer were stratified into a separate cohort (cohort 48) for outcomes analysis at the time of trial design. Pathology was determined by review of local pathology reports by the study principal investigators.

Adult patients (age ≥ 18) with histologically confirmed gallbladder cancer who progressed following at least one line of systemic therapy and who did not have an approved or standard therapy available that had been shown to prolong OS were eligible to enroll. Patients were required to have a Eastern Cooperative Oncology Group (ECOG) performance status of 0–2, measurable disease [as defined in section 10.1 of the revised Response Evaluation Criteria in Solid Tumors (RECIST) v1.124], and adequate hematologic, hepatic, thyroid, adrenal axis, and renal function, with an absolute neutrophil count ≥ 1,000/mcL, platelets ≥ 75,000/mcL, hemoglobin ≥ 8 g/dL, creatinine clearance ≥ 50 mL/minute, total bilirubin ≤ 2.0 × institutional upper limit of normal, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) ≤ 3.0 × institutional upper limit of normal, thyroid-stimulating hormone (TSH) or free T4 serum ≤ institutional upper limit of normal, and normal adrenocorticotropic hormone (ACTH) ≤ institutional upper limit of normal. Women of childbearing potential were required to have a negative serum pregnancy test and subjects were required to practice adequate birth control during protocol participation. Patients may have received anti-CTLA4 or prior anti-PD-(L)1 therapy, but not both, provided that it was completed ≥ 4 weeks prior to registration. Patients who had a grade 3 or higher immune-related adverse event (e.g. pneumonitis, hepatitis, colitis, endocrinopathy) with prior immunotherapy (e.g. cancer vaccine, cytokine, etc.) were not eligible.

Treatment and monitoring

Treatment consisted of nivolumab 240 mg intravenously every 2 weeks and ipilimumab 1 mg/kg intravenously every 6 weeks on a continuous schedule. Treatment was continued until any of the following occurred: disease progression, symptomatic deterioration, treatment delay for any reason >56 days, unacceptable or immune-related toxicity with an inability to decrease prednisone to < 10 mg daily, or per patient request. Dose holds were allowed as specified in the protocol for treatment-related toxicities. Dose reductions were not allowed.

Patients were evaluated with a history and physical, laboratory analyses (complete blood count, comprehensive metabolic panel, TSH, free thyroxine, ACTH, cortisol, lipase), and toxicity assessment at least every 6 weeks at the beginning of each cycle. Imaging studies for disease assessment were performed before treatment began, week 8, week 16, week 24, and then every 12 weeks until progression.

Statistical Analysis and Outcome Endpoints

The primary endpoint was confirmed ORR [complete response (CR) and partial response (PR) confirmed on subsequent scan]; by RECIST v1.1 based on local site review. The design was selected to distinguish between an ORR <5% (null hypothesis, as patients had failed all known active therapies) versus ≥30% (alternative hypothesis, a potentially clinically meaningful difference in tumor response in refractory solid tumors). A two-stage design was used; in the first stage if 1 or more of the 6 patients had a response (confirmed CR or PR), an additional 10 patients were to be accrued in a second stage. If ≥ 2 out of 16 patients had a response, the null hypothesis would be rejected (one-sided alpha = 13%, power = 87%). This cohort accrued more than 16 patients because, unexpectedly, accrual was faster than expected following the 2-week closure notification.

Secondary endpoints included unconfirmed ORR, clinical benefit rate [CR + PR (confirmed + unconfirmed) + stable disease > 6 months], PFS, OS, ORR by immune-related RECIST (iRECIST)25, PFS by iRECIST, and toxicity assessment. PFS was measured from the start of protocol therapy to the first date of progression or death by any cause, with patients last known to be alive without progression censored at the date of last contact. OS was measured from the date of study registration to the date of death by any cause, with patients last known to be alive censored at the date of last contact. PFS and OS estimates were calculated using the Kaplan–Meier method. Confidence intervals for medians were constructed using the method of Brookmeyer and Crowley26, and confidence intervals for point estimates (e.g., 6-month PFS) were calculated using the log–log transformation. Fisher’s exact test was used to compare subgroups. All analyses were performed using R version 4.2.3.

Results

Patient characteristics

Twenty-two patients were registered between October, 2017 and June, 2020 (Figure 1). Two patients did not meet eligibility criteria after registration (missing pathology reports), and one patient was transitioned to hospice before receiving any protocol therapy. Nineteen patients were enrolled and received therapy. At the time of data cut-off, 17/19 (89%) patients had disease progression or had died. The follow-up for the remaining 2 patients was over 34 months.

Figure 1.

Figure 1.

Study Flow Diagram. PFS, progression-free survival

Patient characteristics are summarized in Table 1. The median age was 60 years (range 40–84). Seventy-nine percent of patients were female. The majority of patients identified as White (79%) and non-Hispanic (95%). MSI results were available for 47% of patients; none had MSI-High disease. The median number of prior lines of therapy was 2 (range 1–6). One patient in the cohort had prior treatment with an immune checkpoint inhibitor. This patient was treated with atezolizumab for 10 months after gemcitabine/cisplatin (2nd-line treatment). A complete list of prior therapies can be viewed in eTable 1.

Table 1.

Patient characteristics [median (min, max) or N (%) reported]

Summary (n=19)
Age, years 60 (40, 84)
Gender
 Female 15 (79)
 Male 4 (21)
Race
 Asian 1 (5)
 Black 3 (16)
 White 15 (79)
Ethnicity
 Hispanic or Latino 1 (5)
 Non-Hispanic 18 (95)
ECOG performance status
 0 6 (32)
 1 11 (58)
 2 2 (11)
Histology
 Adenocarcinoma 17 (90)
 Mixed adenocarcinoma and large cell neuroendocrine carcinoma 1 (5)
 Squamous/Adenosquamous cell carcinoma 1 (5)
Differentiation
 Well 1 (5)
 Moderate to well 2 (11)
 Moderate 5 (26)
 Moderate to poor 3 (16)
 Poor 4 (21)
 Missing 4 (32)
MSI Status
 High 0 (0)
 Stable 9 (47)
 Missing 10 (53)
Median prior lines of therapy 2 (1–6)

ECOG, Eastern Cooperative Oncology Group; MSI, microsatellite instability, TMB, tumor mutational burden

Outcomes

Eighteen of the nineteen patients receiving treatment were evaluable for response by RECIST v1.1; one patient died before post-treatment imaging could be obtained. The confirmed ORR was 16% [CR, n=1 (5%); PR, n=2 (11%)] (Figure 2AB). The patient with a confirmed CR had 3 target lesions totaling 4.5 cm at baseline. There were also 3 patients with an unconfirmed partial response (uPR): 1 patient had surgery to remove all residual disease after achieving an uPR and before confirmatory imaging was completed, 1 patient died from sepsis before confirmatory imaging could be performed, and 1 patient had progressive disease on subsequent imaging after achieving an uPR (eFigure 1 in the supplement). Two patients (11%) had SD < 6 months, and no patients had SD > 6 months. The clinical benefit rate [CR + PR (confirmed and unconfirmed) + stable disease > 6 months] was 32%, matching the unconfirmed ORR. No significant differences were noted when patients were evaluated using iRECIST criteria.

Figure 2.

Figure 2.

Figure 2.

Figure 2.

Figure 2.

Waterfall plot (A), Swimmer’s plot (B), and Kaplan-Meier curves of progression-free survival (C) and overall survival (D). Gray lines in the waterfall plot at −30% and 20% indicate lines for partial response and progression per RECIST v1.1, respectively. Hatched bars in the waterfall plot indicate patients who had death before first assessment (n=1), new lesions at first scan (n=4), and symptomatic deterioration and clinical progression (n=1); these patients are shown as 21% increase indicating progression. PFS, progression-free survival.

The patient who had surgery continues to have no evidence of disease at 34.6+ months from ipilimumab/nivolumab start. This patient had 1 target lesion of 8.3 cm and one non-target lesion prior to treatment. They developed grade 3 immune-related hepatitis 295 days after starting therapy and had to come off treatment at that time. They were then treated with a prolonged steroid regimen, and surgery was performed 132 days later. They are now approaching 2 years after surgery with no evidence of disease.

Biomarker testing for microsatellite stability, tumor mutational burden (TMB), and programmed death-ligand 1 (PD-L1) were not routinely reported. However, all the patients with a confirmed objective response were microsatellite stable (Table 2). Seven patients had TMB results (eTable 2 in the supplement). One of these patients was TMB-high [≥10 mutations/megabase (mut/Mb)], and this patient achieved a PR. Four patients had PD-L1 immunohistochemistry results (eTable 3 in the supplement). All four had PD-L1 staining < 1%; one of these patients had a durable complete response.

Table 2.

Microsatellite stable response by RECIST v1.1, N (%)

Microsatellite stable (n=9)
Confirmed complete response 1 (11)
Confirmed partial response 2 (22)
Unconfirmed partial response 2 (22)
Stable disease < 6 months 1 (11)
Progressive disease 3 (33)
Not evaluable 0 (0)
Confirmed objective response rate 3 (33)
Unconfirmed objective response rate 5 (56)

The median time to response was 4.5 months (range, 1.8–10.9 months) (Figure 2B). The median duration of response was 14.6 months at data cutoff. Two patients had ongoing responses at 34.6+ and 35.1+ months (note the 34.6+ month response was the patient who had complete surgical resection at 14.2 months). Biomarker test results for the latter patient were as follows: microsatellite stable, TMB-low (7 mut/Mb), and PD-L1 immunohistochemical staining of 0%; biomarker testing for the former patient was not available. The median PFS was 1.8 months (95% CI 1.5–13.3 months) with ongoing responses (Figure 2C). The 6-month PFS was 26% (95% CI 12–55%). The median OS was 7.0 months (95% CI 3.9–19.1) (Figure 2D).

Toxicities

Treatment-related adverse events (TRAEs) are summarized in Table 3. Sixteen of the 19 evaluable patients experienced a TRAE of any grade (84%). The most common TRAEs were fatigue (32%), anemia (26%), anorexia (26%), aspartate aminotransferase (AST) elevation (21%), and diarrhea (21%). Nine TRAEs were of grade ≥ 3 severity (47%). AST elevation was the most common grade ≥ 3 toxicity (11%, n=2). There was one death from hepatic failure (5%), possibly treatment-related, but this patient also had bacteremia at the time of death. Dose holds were common (7/19, 37%). One patient (5%) discontinued treatment due to toxicity (grade 3 increase in aspartate aminotransferase).

Table 3.

Treatment related adverse events occurring in >10% of patients (N=19)

Any Grade Grade ≥ 3
Any 16 (84%) 9 (47%)
Serious 6 (32%) 6 (32%)
Led to Discontinuation 1 (5%) 1 (5%)
Lead to Death 1 (5%) 1 (5%)
 Fatigue 6 (32%) 0 (0%)
 Anemia 5 (26%) 1 (5%)
 Anorexia 5 (26%) 0 (0%)
 Aspartate aminotransferase increased 4 (21%) 2 (11%)
 Diarrhea 4 (21%) 1 (5%)
 Nausea 3 (15%) 1 (5%)
 Alkaline phosphatase increased 3 (15%) 0 (0%)
 Hypothyroidism 3 (15%) 0 (0%)
 Rash maculo-papular 3 (15%) 0 (0%)
 Alanine aminotransferase increased 2 (11%) 1 (5%)
 Hypercalcemia 2 (11%) 1 (5%)
 Adrenal insufficiency 2 (11%) 0 (0%)
 Arthralgia 2 (11%) 0 (0%)
 Constipation 2 (11%) 0 (0%)
 Hyperglycemia 2 (11%) 0 (0%)
 Hyperthyroidism 2 (11%) 0 (0%)
 Lymphocyte count decreased 2 (11%) 0 (0%)
 Myalgia 2 (11%) 0 (0%)
 Pruritus 2 (11%) 0 (0%)
Immune-mediated 12 (63%) 4 (21%)
 Aspartate aminotransferase increased 4 (21%) 2 (11%)
 Diarrhea 4 (21%) 1 (5%)
 Hypothyroidism 3 (16%) 0 (0%)
 Rash maculo-papular 3 (16%) 0 (0%)
 Alanine aminotransferase increased 2 (11%) 1 (5%)
 Adrenal insufficiency 2 (11%) 0 (0%)
 Arthralgia 2 (11%) 0 (0%)
 Hyperthyroidism 2 (11%) 0 (0%)
 Pruritus 2 (11%) 0 (0%)
 Blood bilirubin increased 1 (5%) 1 (5%)
 Encephalopathy 1 (5%) 1 (5%)
 Lipase increased 1 (5%) 0 (0%)
 Pneumonitis 1 (5%) 0 (0%)
 Serum amylase increased 1 (5%) 0 (0%)

Immune-related adverse events (irAEs) were experienced by 63% of patients, with 21% developing grade 3–4 toxicity (Table 3). The most common all-grade irAEs were hepatitis (21%), diarrhea/colitis (21%), hypothyroidism (16%), and rash (16%). The most common grade ≥ 3 irAEs were hepatitis (16%), diarrhea/colitis (5%), and encephalopathy (5%). Two of the three patients with grade ≥ 3 hepatitis were also known to have liver metastases and lesions that were obstructing the biliary tracts. One of these patients underwent successful biliary stenting with a marked improvement in the liver function tests within 48 hours. The other patient was septic from a gram-negative bacteremia and had new lesions in the liver suggestive of either disease progression or multiple liver abscesses. This patient ultimately decided to enroll in hospice and died within 24 hours.

Discussion

SWOG 1609 DART was a large multi-institutional phase II trial that was led by the Early Therapeutics and Rare Cancers Committee and opened at more than 1000 sites across the United States with the aim of answering an unmet need for novel therapies in rare cancers. Fifty-three individual cohorts were created, accruing a total of 798 patients. Several of these have now been reported, including high-grade neuroendocrine, non-pancreatic neuroendocrine, angiosarcoma, and metaplastic breast cancer2730. Similar to the current gallbladder cohort, the previously reported rare cohorts showed a subset of patients with exceptional/durable responses.

The purpose of this DART sub-study was to investigate the efficacy of dual checkpoint blockade targeting CTLA-4 and PD-1 in relapsed/refractory advanced gallbladder cancer. At the time this trial was designed, ICIs were not approved for use in this setting. Standard of care treatment was largely restricted to chemotherapy, and there were no randomized phase III trials to provide definitive guidance. ICIs had shown incredible promise in other malignancies, with some patients experiencing a DOR > 24 months even in the setting of metastatic disease31. This study achieved its primary objective, with a confirmed ORR of 16%. All of the patients with a confirmed response were MSS. TRAEs were common (84%), but the toxicity profile was similar to prior studies using combinations of anti-PD-1 and anti-CTLA-432.

These results compare favorably to alternative chemotherapy regimens for advanced gallbladder cancer in the second-line or beyond. While the ABC-06 trial was a positive phase III study with FOLFOX, the ORR was only 5%13. The rate of grade ≥ 3 AEs was also high at 69%. Additional phase II trials with regorafenib or FOLFIRI in the second-line had ORRs of 11% and 0% respectively14,15. This raises the question as to whether dual-checkpoint blockade could be superior to these treatment regimens, and further investigation is needed.

ICIs have also now been studied as subsequent therapies for biliary tract cancer, including anti-PD-1 alone or in combination with lenvatinib and anti-PD-(L)1 in combination with anti-CTLA-41720,22. Of particular relevance was the phase I study of durvalumab (anti-PD-L1) with tremelimumab (anti-CTLA-4) and the phase II trials of nivolumab (anti-PD-1) either alone or in combination with ipilimumab17,20,22. The ORR with durvalumab + tremelimumab was 10.8%. In contrast, the ORR with nivolumab monotherapy was 11%, and this improved to 31% in the gallbladder cancer subgroup treated with ipilimumab and nivolumab in CA209–538. Our study showed similar efficacy with a confirmed ORR of 16%. The variability of these trial results may be attributable to differences in patient populations, but it does also raise the question of whether anti-PD1 could be superior to anti-PD-L1 in this setting and whether there is truly increased efficacy with anti-PD-1 and anti-CTLA-4 versus anti-PD-1 alone.

Since the start of our trial, multiple predictive biomarkers have emerged for ICIs, including TMB, PD-L1 expression, and MSI33,34. We investigated the expression of these markers in the patients for whom the data was available. Seven patients had TMB reported, and one was TMB-high (10 mutations/Mb). This patient attained a PR. PD-L1 did not appear to correlate well with the response to therapy in the four patients that had results available (all PD-L1 ≤1%, 2/4 responded). None of the tumors with microsatellite stability testing were MSI-High, consistent with previous reports of low MSI-High prevalence in patients with gallbladder cancer2,35. It is also worth noting that all of the confirmed responses in our study and CA209–538 were MSS, suggesting that this population does indeed benefit from immune checkpoint inhibition22. Furthermore, patients without MSI-High, high TMB, or PD-L1 ≥ 1% achieved objective responses in our trial, including one patient with a CR.

We acknowledge that the incomplete biomarker reporting is a significant limitation of our study, particularly now that tumor-agnostic approvals exist for patients with refractory disease and high TMB, MSI, and a select group of targetable alterations36,37. Correlative studies are planned, and efforts are underway to analyze and report these findings. Additional limitations include the non-randomized study design, small sample size, and lack of mandated central pathology and imaging review.

The results of TOPAZ-1 and KEYNOTE-966 also raise the question of how and when immune checkpoint inhibitors should be used in the second-line and beyond. Most patients will now be treated with anti-PD-(L)1 in the first-line setting, so will there still be efficacy if nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) are used after immune checkpoint inhibitor exposure? This question is currently unresolved, but efficacy has been seen in other cancer types with a median ORR after progression of 15.2% (range, 0–50%)38. Moreover, one patient in our study was previously treated with immune checkpoint inhibition, and this patient achieved a PR that lasted > 1 year. This modest ORR may still be better than chemotherapy-only options, such as FOLFOX with an ORR of 5%. There will also be patients who are not suitable for chemotherapy for a multitude of reasons in whom the option of ipilimumab and nivolumab could be beneficial. Future studies are needed to clarify the role of immunotherapy in these contexts, and in particular, whether dual checkpoint blockade might be preferred over chemotherapy in the anti-PD-(L)1 refractory setting.

In this multi-institutional phase II basket trial, the combination of ipilimumab and nivolumab demonstrated a confirmed ORR of 16% (unconfirmed response rate = 32%) in patients with advanced gallbladder cancer treated with at least one prior line of therapy. All patients with a confirmed response had MSS tumors. Additionally, 2 out of 3 with unconfirmed PRs had MSS tumors; the remaining patient had an unknown MSI status. Adverse events and irAEs occurred at the expected frequency with no signal of increased toxicity compared to prior reports32. Of special interest, two patients continue to do well at just under three years. The results of this trial are encouraging and provide support for a larger, phase III trial. However, it will be critically important to determine: (1) whether the addition of anti-CTLA-4 provides any benefit beyond what can be accomplished with anti-PD-1 alone and (2) biomarker correlates for response, as deep and durable responses were seen in patients who had MSS tumors with negative PD-L1 and low TMB. If the signal of improved efficacy and lower toxicity compared to multi-drug chemotherapy can be confirmed, there will be an opportunity to raise the standard of care for a subset of patients with gallbladder cancer. Correlative biomarker analyses are underway for patients in this study to better define genomic and immunomic predictors of immunotherapeutic response, in particular for those with MSS gallbladder cancer.

Supplementary Material

Supinfo

Acknowledgements

We thank all the patients who agreed to participate in this trial. We also thank study site PIs at University of Kansas Cancer Center, University of Chicago Comprehensive Cancer Center, UC San Diego Moores Cancer Center, University of Florida Health Cancer Center, UPMC Hillman Cancer Center, University of South Alabama Mitchell Cancer Institute, Delaware/Christiana Care NCORP, Moffitt Cancer Center, Georgia NCORP, UC Davis Comprehensive Cancer Center, University of Michigan Comprehensive Cancer Center, Roswell Park Cancer Institute, University of Mississippi Medical Center, New Mexico Minority Underserved NCORP, and Oregon Health & Science University.

This research was supported by NIH/NCI grants U10CA180888, U010CA180819, U10CA180821, U10CA180868 and in part by Bristol-Myers Squibb Company. RK is funded in part by 5U01CA180888-08 and 5UG1CA233198-05. The funding source had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.

Conflict of Interest Statement:

Dr. Patel reported grants or contracts from Amgen, AstraZeneca/MedImmune, Bristol-Myers Squibb, Eli Lilly, Fate Therapeutics, Gilead, Iovance, Merck, Pfizer, Roche/Genentech, SQZ Biotechnologies; consulting fees from Amgen, AstraZeneca, Bristol-Myers Squibb, Certis, Eli Lilly, Jazz, Genentech, Illumina, Merck, Pfizer, Rakuten, Tempus

Dr. Chae reported research grants from Research Grant: Abbvie, Bristol-Myers Squibb (BMS), Biodesix, Freenome, Predicine; honoraria from Roche/Genentech, AstraZeneca, Foundation Medicine, Neogenomics, Guardant Health, Boehringher Ingelheim, Biodesix, Immuneoncia, Lilly Oncology, Merck, Takeda, Lunit, Jazz Pharmaceutical, Tempus, BMS, Regeneron, NeoImmunTech, Esai; participating in Advisory Boards for Roche/Genentech, AstraZeneca, Foundation Medicine, Neogenomics, Guardant Health, Boehringher Ingelheim, Biodesix, Immuneoncia, Lilly Oncology, Merck, Takeda, Lunit, Jazz Pharmaceutical, Tempus, BMS, Regeneron, NeoImmunTech, Esai

Dr. Dennis reported consulting fees from MJH Life Sciences

Dr. Liao reported consulting fees from Incyte, AstraZeneca, Lantheus, Eli Lilly, Transthera, Ipsen, BluePrints Medicine, Genentech, QED, Histosonics, Exelixis; payment or Honoria from OncLive

Dr. Ferri reported serving as board member for Beaver County Cancer and Heart Association; serving as president of Beaver County Medical Society

Dr. George reported Consultation and Scientific Advisory Board for Tempus Labs; Consultation and Scientific Advisory Board for BillionToOne; consultation for Pfizer/Array

Dr. Ryan reported grants or contracts (payment to institution) from Ayala, Bristol-Meyer Squibb, Daiichi-Sankyo, Deciphera, Exelixis, Genentech, Novartis, Karyopharm Therapeutics, Merck, Nektar, Pfizer, Xynomic, PF Argentum IP Holdings LLC, Rain Therapeutics, Shasqi, PTC Therapeutics, NiKang Therapeutics; consulting fees from Synox, Daiichi Sankyo, AVEO, Exelixis, Astra Zeneca, Bristol-Meyer Squibb; Payment for expert testimony from Pfizer, GSK, Boehringer Ingelheim (Payments to him by representative counsel)

Dr. Othus reported support from grant award NIH/NCI/NCTN U010CA180819; consulting fee from Merck, Biosight; participation on a Data Safety Monitoring Board or Advisory Board for Celgene, Glycomimetics, Grifols

Dr. Blanke reported support from National Institutes of Health

Dr. Kurzrock reported research funding from Boehringer Ingelheim, Debiopharm, Foundation Medicine, Genentech, Grifols, Guardant, Incyte, Konica Minolta, Medimmune, Merck Serono, Omniseq, Pfizer, Sequenom, Takeda, and TopAlliance and from the NCI; as well as consultant and/or speaker fees and/or advisory board/consultant for Actuate Therapeutics, AstraZeneca, Bicara Therapeutics, Inc., Biological Dynamics, Caris, Datar Cancer Genetics, Daiichi, EISAI, EOM Pharmaceuticals, Iylon, LabCorp, Merck, NeoGenomics, Neomed, Pfizer, Prosperdtx, Regeneron, Roche, TD2/Volastra, Turning Point Therapeutics, X-Biotech; has an equity interest in CureMatch Inc. and IDbyDNA; serves on the Board of CureMatch and CureMetrix, and is a co-founder of CureMatch.

The following authors reported no disclosures: Elizabeth Guadarrama, Benjamin C. Powers, Elad Sharon, Gabby Lopez

Footnotes

Ethics Statement

The trial was conducted by the Early Therapeutics and Rare Cancers committee of the SouthWest Oncology Group (SWOG) Cancer Research Network. The investigational agents were provided by the Cancer Therapy Evaluation Program (CTEP) of the National Cancer Institute (NCI) under an NCI Collaborative Research and Development Agreement with Bristol-Myers Squibb. All study subjects provided their voluntary, written informed consent using a document approved by the institution’s human subject protection committee. The study was conducted in accordance with the Declaration of Helsinki. The protocol and all amendments were approved by SWOG, the NCI, the NCI central institutional review board, and the regulatory committees at the participating institutions (IRB #170321). All authors had access to the study data and reviewed and approved the final manuscript.

Clinical Trial Registration: NCT02834013 (ClincialTrials.gov)

Data Availability Statement

De-identified data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions.

References

  • 1.Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
  • 2.Valle JW, Kelley RK, Nervi B, Oh DY, Zhu AX. Biliary tract cancer. The Lancet. 2021;397(10272):428–444. doi: 10.1016/S0140-6736(21)00153-7 [DOI] [PubMed] [Google Scholar]
  • 3.Valle J, Wasan H, Palmer DH, et al. Cisplatin plus Gemcitabine versus Gemcitabine for Biliary Tract Cancer. N Engl J Med. 2010;362(14):1273–1281. doi: 10.1056/NEJMoa0908721 [DOI] [PubMed] [Google Scholar]
  • 4.Oh DY, Ruth He A, Qin S, et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced Biliary Tract Cancer. NEJM Evid. 2022;1(8). doi: 10.1056/EVIDoa2200015 [DOI] [PubMed] [Google Scholar]
  • 5.Kelley RK, Ueno M, Yoo C, et al. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. Published online April 2023:S0140673623007274. doi: 10.1016/S0140-6736(23)00727-4 [DOI] [PubMed] [Google Scholar]
  • 6.Abou-Alfa GK, Sahai V, Hollebecque A, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21(5):671–684. doi: 10.1016/S1470-2045(20)30109-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhu AX, Macarulla T, Javle MM, et al. Final Overall Survival Efficacy Results of Ivosidenib for Patients With Advanced Cholangiocarcinoma With IDH1 Mutation: The Phase 3 Randomized Clinical ClarIDHy Trial. JAMA Oncol. 2021;7(11):1669. doi: 10.1001/jamaoncol.2021.3836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Weinberg BA, Xiu J, Lindberg MR, et al. Molecular profiling of biliary cancers reveals distinct molecular alterations and potential therapeutic targets. J Gastrointest Oncol. 2019;10(4):652–662. doi: 10.21037/jgo.2018.08.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Javle MM, Catenacci D, Jain A, et al. Precision medicine for gallbladder cancer using somatic copy number amplifications (SCNA) and DNA repair pathway gene alterations. J Clin Oncol. 2017;35(15_suppl):4076–4076. doi: 10.1200/JCO.2017.35.15_suppl.4076 [DOI] [Google Scholar]
  • 10.Sicklick JK, Fanta PT, Shimabukuro K, Kurzrock R. Genomics of gallbladder cancer: the case for biomarker-driven clinical trial design. Cancer Metastasis Rev. 2016;35(2):263–275. doi: 10.1007/s10555-016-9602-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Harding JJ, Fan J, Oh DY, et al. Zanidatamab for HER2-amplified, unresectable, locally advanced or metastatic biliary tract cancer (HERIZON-BTC-01): a multicentre, single-arm, phase 2b study. Lancet Oncol. 2023;24(7):772–782. doi: 10.1016/S1470-2045(23)00242-5 [DOI] [PubMed] [Google Scholar]
  • 12.Nakamura Y, Mizuno N, Sunakawa Y, et al. Tucatinib and Trastuzumab for Previously Treated Human Epidermal Growth Factor Receptor 2–Positive Metastatic Biliary Tract Cancer (SGNTUC-019): A Phase II Basket Study. J Clin Oncol. Published online September 26, 2023:JCO.23.00606. doi: 10.1200/JCO.23.00606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lamarca A, Palmer DH, Wasan HS, et al. Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial. Lancet Oncol. 2021;22(5):690–701. doi: 10.1016/S1470-2045(21)00027-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sun W, Patel A, Normolle D, et al. A phase 2 trial of regorafenib as a single agent in patients with chemotherapy-refractory, advanced, and metastatic biliary tract adenocarcinoma. Cancer. 2019;125(6):902–909. doi: 10.1002/cncr.31872 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Caparica R, Lengelé A, Bekolo W, Hendlisz A. FOLFIRI as second-line treatment of metastatic biliary tract cancer patients. Autopsy Case Rep. 2019;9(2):e2019087. doi: 10.4322/acr.2019.087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yoo C, Kim K pyo, Jeong JH, et al. Liposomal irinotecan plus fluorouracil and leucovorin versus fluorouracil and leucovorin for metastatic biliary tract cancer after progression on gemcitabine plus cisplatin (NIFTY): a multicentre, open-label, randomised, phase 2b study. Lancet Oncol. 2021;22(11):1560–1572. doi: 10.1016/S1470-2045(21)00486-1 [DOI] [PubMed] [Google Scholar]
  • 17.Kim RD, Chung V, Alese OB, et al. A Phase 2 Multi-institutional Study of Nivolumab for Patients With Advanced Refractory Biliary Tract Cancer. JAMA Oncol. 2020;6(6):888. doi: 10.1001/jamaoncol.2020.0930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lwin Z, Gomez-Roca C, Saada-Bouzid E, et al. LBA41 LEAP-005: Phase II study of lenvatinib (len) plus pembrolizumab (pembro) in patients (pts) with previously treated advanced solid tumours. Ann Oncol. 2020;31:S1170. doi: 10.1016/j.annonc.2020.08.2271 [DOI] [Google Scholar]
  • 19.Piha-Paul SA, Oh D, Ueno M, et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: Results from the keynote −158 and keynote −028 studies. Int J Cancer. 2020;147(8):2190–2198. doi: 10.1002/ijc.33013 [DOI] [PubMed] [Google Scholar]
  • 20.Doki Y, Ueno M, Hsu C, et al. Tolerability and efficacy of durvalumab, either as monotherapy or in combination with tremelimumab, in patients from Asia with advanced biliary tract, esophageal, or head-and-neck cancer. Cancer Med. 2022;11(13):2550–2560. doi: 10.1002/cam4.4593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Marabelle A, Le DT, Ascierto PA, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair–Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol. 2020;38(1):1–10. doi: 10.1200/JCO.19.02105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Klein O, Kee D, Nagrial A, et al. Evaluation of Combination Nivolumab and Ipilimumab Immunotherapy in Patients With Advanced Biliary Tract Cancers: Subgroup Analysis of a Phase 2 Nonrandomized Clinical Trial. JAMA Oncol. 2020;6(9):1405. doi: 10.1001/jamaoncol.2020.2814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.DeSantis CE, Kramer JL, Jemal A. The burden of rare cancers in the United States: The Burden of Rare Cancers in the United States. CA Cancer J Clin. 2017;67(4):261–272. doi: 10.3322/caac.21400 [DOI] [PubMed] [Google Scholar]
  • 24.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026 [DOI] [PubMed] [Google Scholar]
  • 25.Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18(3):e143–e152. doi: 10.1016/S1470-2045(17)30074-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Brookmeyer R, Crowley J. A k -Sample Median Test for Censored Data. J Am Stat Assoc. 1982;77(378):433–440. doi: 10.1080/01621459.1982.10477829 [DOI] [Google Scholar]
  • 27.Patel SP, Mayerson E, Chae YK, et al. A phase II basket trial of Dual Anti–CTLA–4 and Anti–PD–1 Blockade in Rare Tumors (DART) SWOG S1609: High-grade neuroendocrine neoplasm cohort. Cancer. 2021;127(17):3194–3201. doi: 10.1002/cncr.33591 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Patel SP, Othus M, Chae YK, et al. A Phase II Basket Trial of Dual Anti–CTLA-4 and Anti–PD-1 Blockade in Rare Tumors (DART SWOG 1609) in Patients with Nonpancreatic Neuroendocrine Tumors. Clin Cancer Res. 2020;26(10):2290–2296. doi: 10.1158/1078-0432.CCR-19-3356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Adams S, Othus M, Patel SP, et al. A Multicenter Phase II Trial of Ipilimumab and Nivolumab in Unresectable or Metastatic Metaplastic Breast Cancer: Cohort 36 of Dual Anti–CTLA-4 and Anti–PD-1 Blockade in Rare Tumors (DART, SWOG S1609). Clin Cancer Res. 2022;28(2):271–278. doi: 10.1158/1078-0432.CCR-21-2182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wagner MJ, Othus M, Patel SP, et al. Multicenter phase II trial (SWOG S1609, cohort 51) of ipilimumab and nivolumab in metastatic or unresectable angiosarcoma: a substudy of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors (DART). J Immunother Cancer. 2021;9(8):e002990. doi: 10.1136/jitc-2021-002990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, Activity, and Immune Correlates of Anti–PD-1 Antibody in Cancer. N Engl J Med. 2012;366(26):2443–2454. doi: 10.1056/NEJMoa1200690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhou X, Yao Z, Bai H, et al. Treatment-related adverse events of PD-1 and PD-L1 inhibitor-based combination therapies in clinical trials: a systematic review and meta-analysis. Lancet Oncol. 2021;22(9):1265–1274. doi: 10.1016/S1470-2045(21)00333-8 [DOI] [PubMed] [Google Scholar]
  • 33.Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol. 2017;14(11):655–668. doi: 10.1038/nrclinonc.2017.88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Patel SP, Kurzrock R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol Cancer Ther. 2015;14(4):847–856. doi: 10.1158/1535-7163.MCT-14-0983 [DOI] [PubMed] [Google Scholar]
  • 35.Goeppert B, Roessler S, Renner M, et al. Low frequency of mismatch repair deficiency in gallbladder cancer. Diagn Pathol. 2019;14(1):36. doi: 10.1186/s13000-019-0813-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Marcus L, Fashoyin-Aje LA, Donoghue M, et al. FDA Approval Summary: Pembrolizumab for the Treatment of Tumor Mutational Burden–High Solid Tumors. Clin Cancer Res. 2021;27(17):4685–4689. doi: 10.1158/1078-0432.CCR-21-0327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tateo V, Marchese PV, Mollica V, Massari F, Kurzrock R, Adashek JJ. Agnostic Approvals in Oncology: Getting the Right Drug to the Right Patient with the Right Genomics. Pharmaceuticals. 2023;16(4):614. doi: 10.3390/ph16040614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Inno A, Roviello G, Ghidini A, et al. Rechallenge of immune checkpoint inhibitors: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 2021;165:103434. doi: 10.1016/j.critrevonc.2021.103434 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supinfo

Data Availability Statement

De-identified data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions.

RESOURCES