Brain radiotherapy, tremelimumab-mediated CTLA-4-directed blockade +/- trastuzumab in patients with breast cancer brain metastases

David B Page, Kathryn Beal, Stefanie N Linch, Kateri J Spinelli, Micaela Rodine, Darragh Halpenny, Shanu Modi, Sujata Patil, Robert J Young, Thomas Kaley, Taha Merghoub, David Redmond, Phillip Wong, Christopher A Barker, Adi Diab, Larry Norton, Heather L McArthur, David B Page, Kathryn Beal, Stefanie N Linch, Kateri J Spinelli, Micaela Rodine, Darragh Halpenny, Shanu Modi, Sujata Patil, Robert J Young, Thomas Kaley, Taha Merghoub, David Redmond, Phillip Wong, Christopher A Barker, Adi Diab, Larry Norton, Heather L McArthur

Abstract

Breast cancer brain metastases (BCBM) are a common and devastating complication of metastatic breast cancer with conventional systemic therapies demonstrating limited effectiveness. Consequently, radiotherapy (RT) ± surgery remains the cornerstone of BCBM management. Because preclinical and clinical evidence indicate that immune checkpoint blockade (ICB) may synergize with RT to promote systemic tumor regression, we explored the safety and efficacy of RT and concurrent tremelimumab-mediated cytotoxic T-lymphocyte associated protein 4 (CTLA-4) ICB with tremelimumab ± HER2-directed therapy with trastuzumab for BCBM. Eligible patients had BCBM indicated for brain RT. A Simon two-stage design was adopted to evaluate the efficacy of tremelimumab and RT in 20 patients with human epidermal growth factor receptor normal (HER2-) BCBM. The safety of concurrent RT, tremelimumab, and trastuzumab was evaluated in a cohort of 6 HER2+ patients. The primary endpoint was 12-week non-central nervous system (CNS) disease control rate (DCR). Secondary endpoints included safety, survival, and CNS response. Exploratory correlatives included characterization of peripheral blood immune responses among exceptional responders. Tremelimumab plus RT ± trastuzumab was tolerated with no treatment-related grade 4 adverse events reported. The 12-week non-CNS DCR was 10% (2/20) in the HER2- cohort and 33% (2/6) in the HER2+ cohort. One patient with HER2+ disease experienced a durable partial response with evidence of peripheral T-cell activation. Thus, tremelimumab and RT ± trastuzumab was tolerated. Although modest clinical activity was observed in the HER2- efficacy cohort, encouraging responses were observed in the HER2+ safety cohort. Consequently, a trial to determine efficacy in HER2+ BCBM is planned.Clinical Trial Registration Number: NCT02563925.

Conflict of interest statement

D.B.P. has served on the advisory board for Merck, Syndax, Peregrine, Bristol-Myers Squibb, Puma, and Nanostring; has received research support from Bristol-Myers Squibb, Merck, and Brooklyn ImmunoTherapeutics; and has served on the speaker’s bureau for Genentech and Novartis. K.B., K.J.S., M.R., D.H., S.P., T.K., D.R., and C.A.B. have no disclosures. S.N.L. has received patent royalties from Galectin Therapeutics. R.J.Y. has consulted for Agios, Puma, ICON plc and NordicNeuroLab; has received research support from Agios. S.M. has research funding from Genentech, Daiichi Sankyo, Novartis, AstraZeneca, and Seattle Genetics; she has received speaking fees from Genentech, Daiichi Sankyo, AstraZeneca, and Seattle Genetics; she has consulted for Genentech, Daiichi Sankyo, AstraZeneca, Macrogenics, and Seattle Genetics. T.M. is a cofounder and holds an equity in IMVAQ Therapeutics. He is a consultant of Immunos Therapeutics and Pfizer. He has research support from Bristol-Myers Squibb; Surface Oncology; Kyn Therapeutics; Infinity Pharmaceuticals, Inc.; Peregrine Pharmaceuticals, Inc.; Adaptive Biotechnologies; Leap Therapeutics, Inc.; and Aprea. He has patents on applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neo antigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4. P.W. has consulted for Sellas Life Sciences and Leap Therapeutics. A.D. has consultaed for Bristol-Myers Squibb, NEKTAR therapeutics, Pfizer, and Apexigen and has received research support from Bristol-Myers Squibb, Nektar therapeutics, Idera, and Merck. H.L.M. has consulted for Amgen, Bristol-Myers Squibb, Celgene, Eli Lilly, Genentech/Roche, Immunomedics, Merck, OBI Pharma, Pfizer, Puma, Spectrum Pharmaceuticals, Syndax Pharmaceuticals, Peregrine, Calithera, Daiichi Sankyo, Seattle Genetics, AstraZeneca and TapImmune; and has received research support from Bristol-Myers Squibb; MedImmune, LLC/AstraZeneca; BTG; and Merck.

© 2022. The Author(s).

Figures

Fig. 1. Trial schema and overall survival.
Fig. 1. Trial schema and overall survival.
A Trial design showing Simon two-stage design for the HER2− cohort and the safety evaluation for the HER2+ cohort. Three or more DCR responses in 17 patients at the end of the study for the treatment to be considered meritorious for further investigation. However, three additional subjects enrolled in the HER2− cohort to expand sample size for estimation of DCR at week 12. These subjects were not included in the pre-specified primary efficacy determination. B Median OS for HER2− cohort = 4.9 months, median OS for HER2+ cohort = 7.98 months. C Median OS for patients with PD = 4.47 months, median OS for patients with SD or PR = 14.58 months.
Fig. 2. Swimmer’s plot.
Fig. 2. Swimmer’s plot.
The Swimmer’s plot illustrates time on treatment in blue and green for the HER2+ and HER2- cohorts, respectively. Time of death is indicated by the red diamonds.
Fig. 3. HER2+ responder treatment schedule and…
Fig. 3. HER2+ responder treatment schedule and radiological response.
A Prior therapies. B Treatment timeline and blood draws (red) for the patient who responded to tremelimumab (blue) and WBRT (black rectangle) with trastuzumab (gold), showing when she responded (green) to when she relapsed (pink). C Baseline (left) and week 12 (right) PET/CT demonstrating a non-CNS PR in a woman treated with concurrent tremelimumab and WBRT with HER2-directed therapy that was durable at 6 months. H trastuzumab, P pertuzumab, cape capecitabine, T-DM1 trastuzumab emtansine, T paclitaxel, treme tremelimumab, RTradiotherapy.
Fig. 4. Immunologic correlates in the blood…
Fig. 4. Immunologic correlates in the blood of a HER2+ responder.
Peripheral blood mononuclear cells were stained for various markers and analyzed using flow cytometry at weeks 0, 2, 4, 24, and 60 post-treatment initiation (n = 2, technical replicates). A Expression of CD8 (red), FoxP3−CD4 conventional T cells (Tconv; light blue), and FoxP3+CD4 regulatory T cells (Treg; dark blue) out of total CD3+ live cells. B CD8:Treg ratio. (C-F) CD8 (red triangles), CD4 Tconv (light blue circles), and Treg (dark blue squares) were analyzed for expression of (C) Ki-67, (D) ICOS, (E) CTLA-4, and (F) PD-1. G Tim-3 expression on PD-1+ compared to PD-1− CD8 T cells. H Percentage of CD14+ and HLA-DRlo monocytes over time.

References

    1. Brufsky AM, et al. Central nervous system metastases in patients with HER2-positive metastatic breast cancer: incidence, treatment, and survival in patients from registHER. Clin. Cancer Res. 2011;17:4834–4843. doi: 10.1158/1078-0432.CCR-10-2962.
    1. Altaha R, Crowell E, Hobbs G, Higa G, Abraham J. Increased risk of brain metastases in patients with HER-2/neu-positive breast carcinoma. Cancer. 2005;103:442–443. doi: 10.1002/cncr.20813.
    1. Bartsch R, et al. Predicting for activity of second-line trastuzumab-based therapy in her2-positive advanced breast cancer. BMC Cancer. 2009;9:367. doi: 10.1186/1471-2407-9-367.
    1. Frisk G, et al. Incidence and time trends of brain metastases admissions among breast cancer patients in Sweden. Br. J. Cancer. 2012;106:1850–1853. doi: 10.1038/bjc.2012.163.
    1. Gori S, et al. Central nervous system metastases in HER-2–positive metastatic breast cancer patients treated with trastuzumab: incidence, survival, and risk factors. Oncologist. 2007;12:766–773. doi: 10.1634/theoncologist.12-7-766.
    1. Cortes J. et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet.396, 1817–1828 (2020).
    1. Vonderheide RH, et al. Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin. Cancer Res. 2010;16:3485–3494. doi: 10.1158/1078-0432.CCR-10-0505.
    1. Ott PA, Hodi FS, Kaufman HL, Wigginton JM, Wolchok JD. Combination immunotherapy: a road map. J. Immunother. Cancer. 2017;5:16. doi: 10.1186/s40425-017-0218-5.
    1. Demaria S, Golden EB, Formenti SC. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol. 2015;1:1325–1332. doi: 10.1001/jamaoncol.2015.2756.
    1. Spiotto, M., Fu, Y. X. & Weichselbaum, R. R. The intersection of radiotherapy and immunotherapy: mechanisms and clinical implications. Sci. Immunol.1:EaaG1266, (2016).
    1. Twyman-Saint Victor C, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520:373–377. doi: 10.1038/nature14292.
    1. Paulson KG, et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat. Commun. 2018;9:3868. doi: 10.1038/s41467-018-06300-3.
    1. Ahmed KA, et al. Clinical outcomes of melanoma brain metastases treated with stereotactic radiosurgery and anti-PD-1 therapy, anti-CTLA-4 therapy, BRAF/MEK inhibitors, BRAF inhibitor, or conventional chemotherapy. Ann. Oncol. 2016;27:2288–2294. doi: 10.1093/annonc/mdw417.
    1. Ahmed KA, et al. Clinical outcomes of melanoma brain metastases treated with stereotactic radiation and anti-PD-1 therapy. Ann. Oncol. 2016;27:434–441. doi: 10.1093/annonc/mdv622.
    1. Qian JM, Yu JB, Kluger HM, Chiang VLS. Timing and type of immune checkpoint therapy affects early radiographic response of melanoma brain metastases to stereotactic radiosurgery. Cancer. 2016;122:3051–3058. doi: 10.1002/cncr.30138.
    1. Kiess AP, et al. Stereotactic radiosurgery for melanoma brain metastases in patients receiving ipilimumab: safety profile and efficacy of combined treatment. Int. J. Radiat. Oncol. Biol. Phys. 2015;92:368–375. doi: 10.1016/j.ijrobp.2015.01.004.
    1. Liakou CI, et al. CTLA-4 blockade increases IFN-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc. Natl Acad. Sci. 2008;105:14987–14992. doi: 10.1073/pnas.0806075105.
    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. Wang W, et al. Biomarkers on melanoma patient T Cells associated with ipilimumab treatment. J. Transl. Med. 2012;10:146. doi: 10.1186/1479-5876-10-146.
    1. Rugo, H. et al. Safety and antitumor activity of pembrolizumab in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer. Clin. Cancer Res. 24, 2804-2811 (2018).
    1. Modi S, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N. Engl. J. Med. 2020;382:610–621. doi: 10.1056/NEJMoa1914510.
    1. Adams S, et al. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of the phase II KEYNOTE-086 study. Ann. Oncol. 2019;30:405–411. doi: 10.1093/annonc/mdy518.
    1. Adams S, et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann. Oncol. 2019;30:397–404. doi: 10.1093/annonc/mdy517.
    1. Schmid P, et al. Abstract 2986: atezolizumab in metastatic TNBC (mTNBC): long-term clinical outcomes and biomarker analyses. Cancer Res. 2017;77:2986–2986.
    1. Dirix LY, et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN solid tumor study. Breast Cancer Res. Treat. 2018;167:671–686. doi: 10.1007/s10549-017-4537-5.
    1. Park S, et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18:160–170. doi: 10.1016/j.ccr.2010.06.014.
    1. Gall VA, et al. Trastuzumab increases HER2 uptake and cross-presentation by dendritic cells. Cancer Res. 2017;77:5374–5383. doi: 10.1158/0008-5472.CAN-16-2774.
    1. Hodi FS, 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. Hiniker SM, et al. A prospective clinical trial combining radiation therapy with systemic immunotherapy in metastatic melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2016;96:578–588. doi: 10.1016/j.ijrobp.2016.07.005.
    1. Koller KM, et al. Improved survival and complete response rates in patients with advanced melanoma treated with concurrent ipilimumab and radiotherapy versus ipilimumab alone. Cancer Biol. Ther. 2017;18:36–42. doi: 10.1080/15384047.2016.1264543.
    1. Postow MA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012;366:925–931. doi: 10.1056/NEJMoa1112824.
    1. Ngwa W, et al. Using immunotherapy to boost the abscopal effect. Nat. Rev. Cancer. 2018;18:313–322. doi: 10.1038/nrc.2018.6.
    1. Liang K, et al. Sensitization of breast cancer cells to radiation by trastuzumab. Mol. Cancer Therapeutics. 2003;2:353–360.
    1. Pietras RJ, et al. Monoclonal antibody to HER-2/neureceptor modulates repair of radiation-induced DNA damage and enhances radiosensitivity of human breast cancer cells overexpressing this oncogene. Cancer Res. 1999;59:1347–1355.
    1. Fourcade J, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen–specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 2010;207:2175–2186. doi: 10.1084/jem.20100637.
    1. Jin HT, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc. Natl Acad. Sci. USA. 2010;107:14733–14738. doi: 10.1073/pnas.1009731107.
    1. Oweida A, et al. Resistance to radiotherapy and PD-L1 blockade is mediated by TIM-3 upregulation and regulatory T-cell infiltration. Clin. Cancer Res. 2018;24:5368–5380. doi: 10.1158/1078-0432.CCR-18-1038.
    1. Kim JE, et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin. Cancer Res. 2017;23:124–136. doi: 10.1158/1078-0432.CCR-15-1535.
    1. Murthy RK, et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N. Engl. J. Med. 2020;382:597–609. doi: 10.1056/NEJMoa1914609.
    1. Freedman RA, et al. TBCRC 022: a phase II trial of neratinib and capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases. J. Clin. Oncol. 2019;37:1081–1089. doi: 10.1200/JCO.18.01511.
    1. Verma S, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 2012;367:1783–1791. doi: 10.1056/NEJMoa1209124.
    1. Loi S, et al. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): a single-arm, multicentre, phase 1b–2 trial. Lancet Oncol. 2019;20:371–382. doi: 10.1016/S1470-2045(18)30812-X.
    1. Dovedi SJ, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74:5458–5468. doi: 10.1158/0008-5472.CAN-14-1258.
    1. Young, K. H. et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS ONE11:e0157164 (2016).
    1. Young KH, et al. TGF inhibition prior to hypofractionated radiation enhances efficacy in preclinical models. Cancer Immunol. Res. 2014;2:1011–1022. doi: 10.1158/2326-6066.CIR-13-0207.
    1. Vanpouille-Box C, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017;8:15618. doi: 10.1038/ncomms15618.
    1. Gameiro SR, et al. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. 2014;5:403–416. doi: 10.18632/oncotarget.1719.
    1. Morisada M, et al. PD-1 blockade reverses adaptive immune resistance induced by high-dose hypofractionated but not low-dose daily fractionated radiation. Oncoimmunology. 2018;7:e1395996. doi: 10.1080/2162402X.2017.1395996.
    1. Wolchok JD, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 2009;15:7412–7420. doi: 10.1158/1078-0432.CCR-09-1624.
    1. Wen, P. Y. et al. Response assessment in neuro-oncology clinical trials. J. Clin. Oncol.35, 2439-2449 (2017).
    1. Ivanova A, Qaqish BF, Schell MJ. Continuous toxicity monitoring in phase II trials in oncology. Biometrics. 2005;61:540–545. doi: 10.1111/j.1541-0420.2005.00311.x.
    1. Page DB, et al. Deep sequencing of T-cell receptor DNA as a biomarker of clonally expanded TILs in breast cancer after immunotherapy. Cancer Immunol. Res. 2016;4:835–844. doi: 10.1158/2326-6066.CIR-16-0013.

Source: PubMed

3
Se inscrever