Pilot study of Tremelimumab with and without cryoablation in patients with metastatic renal cell carcinoma

Matthew T Campbell, Surena F Matin, Alda L Tam, Rahul A Sheth, Kamran Ahrar, Rebecca S Tidwell, Priya Rao, Jose A Karam, Christopher G Wood, Nizar M Tannir, Eric Jonasch, Jianjun Gao, Amado J Zurita, Amishi Y Shah, Sonali Jindal, Fei Duan, Sreyashi Basu, Hong Chen, Alexsandra B Espejo, James P Allison, Shalini S Yadav, Padmanee Sharma, Matthew T Campbell, Surena F Matin, Alda L Tam, Rahul A Sheth, Kamran Ahrar, Rebecca S Tidwell, Priya Rao, Jose A Karam, Christopher G Wood, Nizar M Tannir, Eric Jonasch, Jianjun Gao, Amado J Zurita, Amishi Y Shah, Sonali Jindal, Fei Duan, Sreyashi Basu, Hong Chen, Alexsandra B Espejo, James P Allison, Shalini S Yadav, Padmanee Sharma

Abstract

Cryoablation in combination with immune checkpoint therapy was previously reported to improve anti-tumor immune responses in pre-clinical studies. Here we report a pilot study of anti-CTLA-4 (tremelimumab) with (n = 15) or without (n = 14) cryoablation in patients with metastatic renal cell carcinoma (NCT02626130), 18 patients with clear cell and 11 patients with non-clear cell histologies. The primary endpoint is safety, secondary endpoints include objective response rate, progression-free survival, and immune monitoring studies. Safety data indicate ≥ grade 3 treatment-related adverse events in 16 of 29 patients (55%) including 6 diarrhea/colitis, 3 hepatitis, 1 pneumonitis, and 1 glomerulonephritis. Toxicity leading to treatment discontinuation occurs in 5 patients in each arm. 3 patients with clear cell histology experience durable responses. One patient in the tremelimumab arm experiences an objective response, the median progression-free survival for all patients is 3.3 months (95% CI: 2.0, 5.3 months). Exploratory immune monitoring analysis of baseline and post-treatment tumor tissue samples shows that treatment increases immune cell infiltration and tertiary lymphoid structures in clear cell but not in non-clear cell. In clear cell, cryoablation plus tremelimumab leads to a significant increase in immune cell infiltration. These data highlight that treatment with tremelimumab plus cryotherapy is feasible and modulates the immune microenvironment in patients with metastatic clear cell histology.

Conflict of interest statement

M.T.C. reports consulting/advisory roles for AstraZeneca, Astellas, Eisai, EMD Serono, Exelixis, Genentech, Pfizer, Seattle Genetics, Consulting: AXDev, Exelixis, Pfizer, Research grants: ApricityHealth, EMD Serono, Exelixis, Janssen, Pfizer, Non-CME education: Bristol Myers-Squibb, Merck, Roche, Pfizer. S.F.M. reports consulting/advisory roles for Johnson and Johnson and Research Funding from QED; Education—Ology education. A.L.T. reports consulting/advisory roles for Boston Scientific, Cello Therapeutics, Endocare, AstraZeneca; and Research Funding from Boston Scientific, Johnson & Johnson. R.A.S. reports consulting/advisory roles for Varian, Boston Scientific. J.A.K. reports consulting/advisory roles for Pfizer, Merck, Roche/Genentech, and Research Funding from Roche/Genentech, Mirati, Merck, Elypta; Stock Ownership: Allogene, MedTek, ROM Technologies. N.M.T. reports consulting/advisory roles for Bristol Myers-Squibb; Pfizer; Nektar Therapeutics; Exelisis, Inc, Eisai Medical Research; Eli Lilly; Oncorena; Calithera Bioscience; Surface Oncology; Novartis, Ipsen; Merck Sharp & Dohme and Research Funding from Bristol Myers-Squibb; Nektar Therapeutics; Calithera Bioscience; Arrowhead Pharmaceuticals, Inc. E.J. reports consulting/advisory roles for Eisai, Exelixis, Novartis, Merck, Pfizer, Roche. And Research support from Exelixis, Merck, Pfizer. J.G. reports consulting/advisory roles for Seattle Genetics, Jounce Therapeutics, Pfizer, Janssen, Symphogen, AstraZeneca, ARMO biosciences, CRISPR Therapeutics AG. A.J.Z. reports consulting/advisory roles for AstraZeneca and Bayer and Research funding to his institution from Infinity Pharma, Honoraria from Pfizer, and Janssen. A.Y.S. reports consulting/advisory roles for Ad boards, Pfizer, Exelixis, and Research Grant funding from Bristol Myers-Squibb, Eisai, EMD Serono. J.P.A. reports consulting, advisory roles, and/or stocks/ownership for Achelois, Apricity Health, BioAtla, Codiak BioSciences, Dragonfly Therapeutics, Forty-Seven Inc., Hummingbird, ImaginAb, Jounce Therapeutics, Lava Therapeutics, Lytix Biopharma, Marker Therapeutics, Polaris, BioNTech, and Adaptive Biotechnologies; and owns a patent licensed to Jounce (61/247,438; “Combination Immunotherapy for the Treatment of Cancer”). P.S. reports consulting, advisory roles, and/or stocks/ownership for Achelois, Apricity Health, BioAlta, Codiak BioSciences, Constellation, Dragonfly Therapeutics, Forty-Seven Inc., Hummingbird, ImaginAb, Jounce Therapeutics, Lava Therapeutics, Lytix Biopharma, Marker Therapeutics, Oncolytics, Infinity Pharma, BioNTech, Adaptive Biotechnologies, and Polaris; and owns a patent licensed to Jounce Therapeutics (61/247,438; “Combination Immunotherapy for the Treatment of Cancer”). The remaining authors declare no potential competing interest.

© 2021. The Author(s).

Figures

Fig. 1. Study design and outcomes.
Fig. 1. Study design and outcomes.
a Clinical trial schema. b Consort diagram showing patients accrued in the study. c and d Kaplan–Meier curves showing estimates of c overall survival and d progression-free survival, orange lines denote Treme alone arm and purple line denotes Cryo+Treme arm (e) Swimmer’s Plot with PFS based on treatment arm. Orange lines denote patients in the Treme alone arm, purple lines denote patients in the Cryo+Treme arm and green triangles show patients with an ongoing response. Treme tremelimumab monotherapy, Cryo+Treme Cryoablation+Tremelimumab combination therapy, PD progressive disease, PFS progression-free survival.
Fig. 2. Durable responses in mRCC patients…
Fig. 2. Durable responses in mRCC patients with clear cell histology.
a Patient #19 with clear cell histology with an ongoing radiographic response showing baseline cross-sectional imaging (2.5 mm cuts) showing lung metastasis (31.5 mm × 27.5 mm) and post-treatment response at 18 months (marked with red line). Radiographic analysis related to the image shown was performed once. b Patient #19 with favorable changes in the tumor immune microenvironment. Single stain IHC images showing an increased infiltration with T cells, B cells, and immune cells expressing Granzyme B (GrB) and PD-1 post treatment with two doses of tremelimumab. The analysis shown in the micrographs was performed once. c Swimmer’s plot with PFS based on histology. Kaplan–Meier curve of overall survival in mccRCC versus mnccRCC patients. Blue lines denote mccRCC patients, red lines denote mnccRCC patients, and green arrows in the swimmer’s plot show patients with ongoing responses. IHC immunohistochemistry, PFS progression-free survival, mccRCC metastatic clear cell renal cell carcinoma, mnccRCC metastatic non-clear cell renal cell carcinoma.
Fig. 3. Patients with mccRCC have a…
Fig. 3. Patients with mccRCC have a favorable tumor immune microenvironment.
a–c Nanostring analysis comparing pre-treatment tissue samples of 20 mRCC patients with clear cell (mccRCC) versus non-clear cell (mnccRCC) histology (mccRCC, n = 11; mnccRCC, n = 9). a Volcano plot illustrating differentially expressed genes (DEGs). Colored points above the cutoff in the plot show DEGs with p < 0.05 and fold change ≥1.5 that are either upregulated (red) or downregulated (blue) in mccRCC compared to mnccRCC samples. Two-sided P values are from Welch’s t test. b Bar plots showing differentially activated pathways in tumor tissue samples from patients with mccRCC versus mnccRCC. Upregulated pathways in mccRCC are indicated by red bars while downregulated pathways are indicated by blue bars. P values (FDR-adjusted <0.2) are from GSEA algorithm and c Boxplot showing infiltration with Th1 cells. The ends of the box are the upper and lower quartiles (75th and 25th percentiles), the median is the horizontal line inside the box, and whisker lines extend to outliers (minimum and maximum). P value was calculated using the two-tailed unpaired Mann–Whitney test. Each patient is identified by a unique symbol as shown in the inset on the right. Red box whiskers show mccRCC and blue box whiskers show mnccRCC cohort of patients. Patients with ongoing responses are shown in green. P < 0.05 denotes significant differences. mccRCC metastatic clear cell renal cell carcinoma, mnccRCC metastatic non-clear cell renal cell carcinoma, NES normalized enrichment score, Th1 T helper cells type 1. Source data are provided as Source Data file.
Fig. 4. Treatment with tremelimumab monotherapy or…
Fig. 4. Treatment with tremelimumab monotherapy or cryo-tremelimumab combination therapy leads to favorable changes in post-treatment tissue samples of patients with clear cell histology.
ad Pre and post-treatment tissue samples of patients with clear cell (mccRCC) and non-clear cell (mnccRCC) histology were analyzed by NanoString (a and c, mccRCC: pre (n = 11), post (n = 11); mnccRCC: pre (n = 9), post (n = 5)) and IHC (b, d and e, mccRCC: pre (n = 13), post (n = 11); mnccRCC: pre (n = 11), post (n = 5)). Box plots showing a infiltration of CD8 T cells, B cells, and cytotoxic cells, b percentage of CD3 T cells, CD8 T cells, CD20+ B cells, and immune cells expressing Granzyme B and PD-1, c TLS gene signature score and, d TLS density. For box plots (ad), the ends of the boxes are the upper and lower quartiles (75th and 25th percentiles), the median is the horizontal line inside the box, and whisker lines extend to outliers (minimum and maximum). Each patient is identified by a unique symbol as shown in the inset on the right. Patients with ongoing responses are shown in green. Red box whiskers show mccRCC and blue box whiskers show mnccRCC cohort of patients. P values were calculated using the two-tailed unpaired Mann–Whitney test. P < 0.05 denotes significant differences. P values were adjusted for multiple comparisons using the Benjamini–Hochberg procedure. e Representative single stain IHC images showing a TLS from a post-treatment tumor tissue sample of patient #2 with clear cell histology who has an ongoing response. The analysis shown in the micrographs was performed once. IHC immunohistochemistry, mccRCC; metastatic clear cell renal cell carcinoma, mnccRCC metastatic non-clear cell renal cell carcinoma, Pre pre-treatment sample, Post post-treatment sample, GrB granzyme B, TLS tertiary lymphoid structures. All source data are provided as a Source Data file.
Fig. 5. Adding cryoablation to tremelimumab leads…
Fig. 5. Adding cryoablation to tremelimumab leads to immune cell infiltration in the tumor microenvironment of mRCC patients with clear cell histology.
Pre- and post-treatment tissue samples from patients with clear cell histology who had evaluable tumor tissue samples were analyzed for immune cell infiltration by IHC. a Box plots showing infiltration of CD3 and CD8 T cells, CD20 + B cells, and immune cells expressing PD-1 in the tumor microenvironment of pre, (red, n = 12) and post-treatment tumor samples of patients treated with tremelimumab monotherapy (post Treme, orange, n = 4) or cryoablation plus tremelimumab combination therapy (Post Cryo+Treme, purple, n = 5). For the box plots, the ends of the box are the upper and lower quartiles (75th and 25th percentiles), the median is the horizontal line inside the box, whisker lines extend to outliers (minimum and maximum) and b Bar plots showing the preferential distribution of immune cells as in a between the tumor and stromal compartments of the tumor microenvironment. Pre shown in red, post Treme shown in orange, and Post Cryo+Treme shown in purple. Patients with ongoing responses are shown in green. Error bars represent the mean with minimum to maximum value range. P values were calculated using a two-tailed, unpaired Mann–Whitney test. P values were adjusted for multiple comparisons using the Benjamini–Hochberg procedure. P < 0.05 denotes significant differences. Pre pre-treatment sample, Post Treme, post-treatment sample treated with tremelimumab monotherapy, Post Cryo+Treme post-treatment sample treated with cryoablation plus tremelimumab combination therapy. All source data are provided as a Source Data file.

References

    1. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271:1734–1736. doi: 10.1126/science.271.5256.1734.
    1. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Sci. 1995;182:459–465. doi: 10.1084/jem.182.2.459.
    1. Hodi SF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466.
    1. Schadendorf D, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J. Clin. Oncol. 2015;33:1889–1894. doi: 10.1200/JCO.2014.56.2736.
    1. Ribas A, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J. Clin. Oncol. 2013;31:616–622. doi: 10.1200/JCO.2012.44.6112.
    1. Liakou CI, et al. CTLA-4 blockade increases IFNγ-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. PNAS. 2008;105:14987–14992. doi: 10.1073/pnas.0806075105.
    1. Chen H, et al. Anti-CTLA-4 therapy results in higher CD4+ICOShi T cell frequency and IFN-γ levels in both nonmalignant and malignant prostate tissues. PNAS. 2009;106:2729–2734. doi: 10.1073/pnas.0813175106.
    1. Carthon BC, et al. Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin. Cancer Res. 2010;16:2861–2871. doi: 10.1158/1078-0432.CCR-10-0569.
    1. Ng Tang D, et al. Increased frequency of ICOS+ CD4 T Cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol. Res. 2013;1:229–234. doi: 10.1158/2326-6066.CIR-13-0020.
    1. Choueiri TK, Motzer RJ. Systemic therapy for metastatic renal-cell carcinoma. N. Engl. J. Med. 2017;376:354–366. doi: 10.1056/NEJMra1601333.
    1. Yang JC, et al. Ipilimumab (Anti-CTLA4 Antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J. Immunother. 2007;30:825–830. doi: 10.1097/CJI.0b013e318156e47e.
    1. Bang HJ, et al. Percutaneous cryoablation of metastatic renal cell carcinoma for local tumor control: feasibility, outcomes, and estimated cost-effectivness for palliation. J. Vasc. Interv. Radiol. 2012;23:770–777. doi: 10.1016/j.jvir.2012.03.002.
    1. Callstrom MR, et al. Percutaneous image-guided cryoablation of painful metastases involving bone: multicenter trial. Cancer. 2013;119:1033–1041. doi: 10.1002/cncr.27793.
    1. Sabel MS. Cryo-immunology: a review of the literature and proposed mechanisms for stimulatory versus suppressive immune responses. Cryobiology. 2008;58:1–11. doi: 10.1016/j.cryobiol.2008.10.126.
    1. Matin SF, et al. Immunological response to renal cryoablation in an in vivo orthotopic renal cell carcinoma murine model. J. Urol. 2010;183:333–338. doi: 10.1016/j.juro.2009.08.110.
    1. Waitz R, Fasso M, Allison JP. CTLA-4 blockade synergizes with cryoablation to mediate tumor rejection. Oncoimmunology. 2012;1:544–546. doi: 10.4161/onci.19442.
    1. Seymour, L. et al. RECIST working group. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 18, 30074–30078 (2017).
    1. Gao J, et al. Neoadjuvant PD-L1 plus CTLA-4 blockade in patients with cisplatin-ineligible operable high-risk urothelial carcinoma. Nat. Med. 2020;26:1845–1851. doi: 10.1038/s41591-020-1086-y.
    1. Helmink BA, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549–555. doi: 10.1038/s41586-019-1922-8.
    1. Cabrita R, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–565. doi: 10.1038/s41586-019-1914-8.
    1. Petitprez F, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556–560. doi: 10.1038/s41586-019-1906-8.
    1. Hammers HJ, et al. Safety and efficacy of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma: the CheckMate 016 study. J. Clin. Oncol. 2017;35:3851–3858. doi: 10.1200/JCO.2016.72.1985.
    1. Motzer RJ, et al. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma. N. Engl. J. Med. 2021;384:1289–1300. doi: 10.1056/NEJMoa2035716.
    1. Choueiri TK, et al. Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 2021;384:829–841. doi: 10.1056/NEJMoa2026982.
    1. Rini BI, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N. Engl. J. Med. 2019;380:1116–1127. doi: 10.1056/NEJMoa1816714.
    1. Motzer RJ, et al. Avelumab plus axitinib versus sunitinib in advanced renal cell carcinoma: biomarker analysis of the phase 3 JAVELIN Renal 101 trial. Nat. Med. 2020;26:1733–1741. doi: 10.1038/s41591-020-1044-8.
    1. Ricketts CJ, et al. The Cancer Genome Atlas comprehensive molecular characterization of renal cell carcinoma. Cell Rep. 2018;23:313–326. doi: 10.1016/j.celrep.2018.03.075.
    1. Ayers M, et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 2017;127:2930–2940. doi: 10.1172/JCI91190.
    1. Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011.
    1. Van Allen EM, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–211. doi: 10.1126/science.aad0095.
    1. Choueiri TK, et al. Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma. Clin. Cancer Res. 2016;22:5461–5471. doi: 10.1158/1078-0432.CCR-15-2839.
    1. McDermott DF, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat. Med. 2018;24:749–757. doi: 10.1038/s41591-018-0053-3.
    1. Basu S, Binder RJ, Suto R, Anderson KM. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-κB pathway. Int. Immunol. 2000;12:1539–1546. doi: 10.1093/intimm/12.11.1539.
    1. Kato T, et al. Characterization of the cryoablation-induced immune response in kidney cancer patients. Oncoimmunology. 2017;6:e1326441. doi: 10.1080/2162402X.2017.1326441.
    1. Thakur A, et al. Induction of specific cellular and humoral responses against renal cell carcinoma after combination therapy with cryoablation and granulocyte-macrophage colony stimulating factor: a pilot study. J. Immunother. 2011;34:457–467. doi: 10.1097/CJI.0b013e31821dcba5.
    1. McArthur HL, et al. A pilot study of preoperative single-dose ipilimumab and/or cryoablation in women with early-stage breast cancer with comprehensive immune profiling. Clin. Cancer Res. 2016;22:5729–5737. doi: 10.1158/1078-0432.CCR-16-0190.
    1. Thall PF, Simon RM, Estey EH. Bayesian sequential monitoring designs for single-arm clinical trials with multiple outcomes. Stat. Med. 1995;14:357–379. doi: 10.1002/sim.4780140404.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.

Source: PubMed

3
S'abonner