Pembrolizumab in combination with gemcitabine for patients with HER2-negative advanced breast cancer: GEICAM/2015-04 (PANGEA-Breast) study

L de la Cruz-Merino, M Gion, J Cruz, J L Alonso-Romero, V Quiroga, F Moreno, R Andrés, M Santisteban, M Ramos, E Holgado, J Cortés, E López-Miranda, A Cortés, F Henao, N Palazón-Carrión, L M Rodriguez, I Ceballos, A Soto, A Puertes, M Casas, S Benito, M Chiesa, S Bezares, R Caballero, C Jiménez-Cortegana, V Sánchez-Margalet, F Rojo, L de la Cruz-Merino, M Gion, J Cruz, J L Alonso-Romero, V Quiroga, F Moreno, R Andrés, M Santisteban, M Ramos, E Holgado, J Cortés, E López-Miranda, A Cortés, F Henao, N Palazón-Carrión, L M Rodriguez, I Ceballos, A Soto, A Puertes, M Casas, S Benito, M Chiesa, S Bezares, R Caballero, C Jiménez-Cortegana, V Sánchez-Margalet, F Rojo

Abstract

Background: We evaluated a new chemoimmunotherapy combination based on the anti-PD1 monoclonal antibody pembrolizumab and the pyrimidine antimetabolite gemcitabine in HER2- advanced breast cancer (ABC) patients previously treated in the advanced setting, in order to explore a potential synergism that could eventually obtain long term benefit in these patients.

Methods: HER2-negative ABC patients received 21-day cycles of pembrolizumab 200 mg (day 1) and gemcitabine (days 1 and 8). A run-in-phase (6 + 6 design) was planned with two dose levels (DL) of gemcitabine (1,250 mg/m2 [DL0]; 1,000 mg/m2 [DL1]) to determine the recommended phase II dose (RP2D). The primary objective was objective response rate (ORR). Tumor infiltrating lymphocytes (TILs) density and PD-L1 expression in tumors and myeloid-derived suppressor cells (MDSCs) levels in peripheral blood were analyzed.

Results: Fourteen patients were treated with DL0, resulting in RP2D. Thirty-six patients were evaluated during the first stage of Simon's design. Recruitment was stopped as statistical assumptions were not met. The median age was 52; 21 (58%) patients had triple-negative disease, 28 (78%) visceral involvement, and 27 (75%) ≥ 2 metastatic locations. Progression disease was observed in 29 patients. ORR was 15% (95% CI, 5-32). Eight patients were treated ≥ 6 months before progression. Fourteen patients reported grade ≥ 3 treatment-related adverse events. Due to the small sample size, we did not find any clear association between immune tumor biomarkers and treatment efficacy that could identify a subgroup with higher probability of response or better survival. However, patients that experienced a clinical benefit showed decreased MDSCs levels in peripheral blood along the treatment.

Conclusion: Pembrolizumab 200 mg and gemcitabine 1,250 mg/m2 were considered as RP2D. The objective of ORR was not met; however, 22% patients were on treatment for ≥ 6 months. ABC patients that could benefit of chemoimmunotherapy strategies must be carefully selected by robust and validated biomarkers. In our heavily pretreated population, TILs, PD-L1 expression and MDSCs levels could not identify a subgroup of patients for whom the combination of gemcitabine and pembrolizumab would induce long term benefit.

Trial registration: ClinicalTrials.gov and EudraCT (NCT03025880 and 2016-001,779-54, respectively). Registration dates: 20/01/2017 and 18/11/2016, respectively.

Keywords: Advanced breast cancer; Chemotherapy; HER2-negative; MDSCs; PD-L1; Pembrolizumab; TILs.

Conflict of interest statement

L. de la Cruz-Merino received consulting or advisory role fees from Merck Sharp & Dohme (MSD)-Merck, Roche, Bristol-Myers-Squibb (BMS), Pierre-Fabre, Amgen, and Novartis; research funding from MSD-Merck, Roche, and Celgene; speaker’s honoraria from MSD-Merck, Roche, BMS, and Amgen; and grant support from Roche and BMS. M Gion received honoraria from Roche and traveled accommodation from Pfizer. J. Cruz received consulting or advisory role fees from Roche, PharmaMar, Lilly, Novartis, Eisai, Pfizer Amgen, and Celgene; travel accommodation from Roche, Novartis, and PharmaMar. V. Quiroga received speakers’ bureau honoraria from Pfizer, Novartis, and Roche; research funding from Celgene; and travel accommodation from Novartis, Roche, and Pfizer. F. Moreno received an advisory role honoraria from Pfizer, Roche, Novartis MSD, and AstraZeneca; speaker’s bureau honoraria and research funding from Pfizer; and travel accommodation from Pfizer, Roche, and Novartis. R. Andrés received travel accommodation from Roche. M. Santisteban received honoraria from Roche, Novartis, and Pfizer; and consulting or advisory role honoraria from Gilead, MSD, Novartis, Pfizer, and Biomerieux. M. Ramos received speakers’ bureau honoraria from Novartis, Roche, and Pfizer. J. Cortés has stock and other ownership interests in MedSIR and has received honoraria from Novartis, Eisai, Celgene, Pfizer, Roche, SAMSUNG, Lilly, MSD, and Daiichi Sankyo; consulting or advisory role honoraria from Celgene, Cellestia Biotech, AstraZeneca, Biothera, Merus, Roche, Seattle Genetics, Daiichi Sankyo, ERYTECH Pharma, Polyphor, Athenex, Lilly, Servier, MSD, GlaxoSmithKline (GSK), Leuko, Clovis Oncology, Bioasis, Boehringer Ingelheim, and Kyowa Kyrin; research funding from ARIAD, AstraZeneca, Baxalta GMBH/Servier Affaires, Bayer, Eisai Farmaceutica, Guardanth health, MSD, Pfizer, Puma CO, Queen Mary University of London, Roche, and Piqur; and travel accommodation from Roche, Pfizer, Eisai, Novartis, and Daiichi Sankyo. E. López received consulting or advisory role honoraria from AstraZeneca, Pfizer, Roche, and Novartis; and the speaker’s bureau honoraria from Roche, Eisai, Pfizer, and Novartis. A. Cortés received consulting or advisory role honoraria from Lilly, Roche, Clovis, Ferrer, and Pfizer; speakers’ bureau honoraria from MSD, GSK, AstraZeneca, Roche and Pfizer, and research funding and travel accommodation from Pfizer. L. M. Rodríguez received honoraria from Pfizer and Pierre Fabre. I. Ceballos received consulting or advisory role honoraria from Roche and Merk KGaA; speakers’ bureau honoraria from Roche, Pfizer, BMS, and Celgene; and travel accommodation from Roche, Merck, Pfizer, and Novartis. F. Rojo received consulting or advisory role fees from Roche, AstraZeneca, Novartis, BMS, MSD, Lilly, Pfizer, Genomic Health, Guardant Health, Archer, and Pierre-Fabre; speaker bureau/expert testimony fees from Roche, AstraZeneca, Novartis, BMS, MSD, Lilly, Pfizer, and Pierre-Fabre; and travel accommodation from Roche and Novartis His institution received research grant/funding from Roche and Pfizer. The rest of authors declare no conflict of interest.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Study flowchart
Fig. 2
Fig. 2
Run-in phase design, patient inclusion, and DLTs
Fig. 3
Fig. 3
Swimmer plot for the intention-to-treat population
Fig. 4
Fig. 4
TILs density distribution according to tumor subtype and OR (yes/no)
Fig. 5
Fig. 5
PD-L1 CPS distribution according to tumor subtype and OR (yes/no)
Fig. 6
Fig. 6
Correlation of TILs density and PD-L1 expression
Fig. 7
Fig. 7
Baseline median values of MDSCs in ABC patients and healthy cohort

References

    1. Administration FaD. KEYTRUDA® (pembrolizumab) prescribing information. 2019. Available from: .
    1. Administration FaD . OPDIVO (nivolumab) prescribing information. 2019.
    1. Administration FaD . TECENTRIQ® (atezolizumab) prescribing informatioin. 2019.
    1. Pusztai L, Karn T, Safonov A, Abu-Khalaf MM, Bianchini G. New strategies in breast cancer: immunotherapy. Clin Cancer Res. 2016;22(9):2105–2110. doi: 10.1158/1078-0432.CCR-15-1315.
    1. Marra A, Viale G, Curigliano G. Recent advances in triple negative breast cancer: the immunotherapy era. BMC Med. 2019;17(1):90. doi: 10.1186/s12916-019-1326-5.
    1. Zou Y, Zou X, Zheng S, Tang H, Zhang L, Liu P, et al. Efficacy and predictive factors of immune checkpoint inhibitors in metastatic breast cancer: a systematic review and meta-analysis. Ther Adv Med Oncol. 2020;12:1758835920940928.
    1. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379(22):2108–2121. doi: 10.1056/NEJMoa1809615.
    1. Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im S-A, Yusof MM, 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. 2020;396(10265):1817–1828. doi: 10.1016/S0140-6736(20)32531-9.
    1. Rugo HS, Schmid P, Cescon DW, Nowecki Z, Im S-A, Yusof MM, et al. Abstract GS3–01: Additional efficacy endpoints from the phase 3 KEYNOTE-355 study of pembrolizumab plus chemotherapy vs placebo plus chemotherapy as first-line therapy for locally recurrent inoperable or metastatic triple-negative breast cancer. Cancer Res. 2021;81((4 Supplement)):GS3-01–GS3. doi: 10.1158/1538-7445.SABCS20-GS3-01.
    1. Miles DAF, Gligorov J, Verma S, Xu B, Cameron D, Barrios CH, Schneeweiss A, Easton V, Dolado I, O’Shaughnessy J. IMpassion131: phase III study comparing 1L atezolizumab with paclitaxel vs placebo with paclitaxel in treatment-naive patients with inoperable locally advanced or metastatic triple negative breast cancer (mTNBC) Ann Oncol. 2017;2017(28):105. doi: 10.1093/annonc/mdx365.080.
    1. Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer 2013. Ann Oncol. 2013;24(9):2206–2223. doi: 10.1093/annonc/mdt303.
    1. Nowak AK, Lake RA, Marzo AL, Scott B, Heath WR, Collins EJ, et al. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells. J Immunol. 2003;170(10):4905–4913. doi: 10.4049/jimmunol.170.10.4905.
    1. Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest. 2006;116(10):2777–2790. doi: 10.1172/JCI28828.
    1. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res. 2005;11(18):6713–6721. doi: 10.1158/1078-0432.CCR-05-0883.
    1. Zhang X, Wang D, Li Z, Jiao D, Jin L, Cong J, et al. Low-dose gemcitabine treatment enhances immunogenicity and natural killer cell-driven tumor immunity in lung cancer. Front Immunol. 2020;11:331. doi: 10.3389/fimmu.2020.00331.
    1. DA Piadel K, Smith PL. Gemcitabine in the era of cancer immunotherapy. J Clin Haematol. 2020;1(4):107–120.
    1. Spielmann M, Llombart-Cussac A, Kalla S, Espié M, Namer M, Ferrero JM, et al. Single-agent gemcitabine is active in previously treated metastatic breast cancer. Oncology. 2001;60(4):303–307. doi: 10.1159/000058524.
    1. Possinger K, Kaufmann M, Coleman R, Stuart NS, Helsing M, Ohnmacht U, et al. Phase II study of gemcitabine as first-line chemotherapy in patients with advanced or metastatic breast cancer. Anticancer Drugs. 1999;10(2):155–162. doi: 10.1097/00001813-199902000-00003.
    1. Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international immunooncology biomarkers working group: part 1: assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv Anat Pathol. 2017;24(5):235–251. doi: 10.1097/PAP.0000000000000162.
    1. Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de Vijver KK, de Maaker M, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med. 2019;25(6):920–928. doi: 10.1038/s41591-019-0432-4.
    1. Schmid P, Cortes J, Pusztai L, McArthur H, Kummel S, Bergh J, et al. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382(9):810–821. doi: 10.1056/NEJMoa1910549.
    1. Safarzadeh E, Hashemzadeh S, Duijf PHG, Mansoori B, Khaze V, Mohammadi A, et al. Circulating myeloid-derived suppressor cells: an independent prognostic factor in patients with breast cancer. J Cell Physiol. 2019;234(4):3515–3525. doi: 10.1002/jcp.26896.
    1. Cortés J, Lipatov O, Im SA, Gonçalves A, Lee KS, Schmid P, et al. LBA21 - KEYNOTE-119: Phase III study of pembrolizumab (pembro) versus single-agent chemotherapy (chemo) for metastatic triple negative breast cancer (mTNBC) Ann Oncol. 2019;30:v859–v860. doi: 10.1093/annonc/mdz394.010.
    1. Hodge JW, Ardiani A, Farsaci B, Kwilas AR, Gameiro SR. The tipping point for combination therapy: cancer vaccines with radiation, chemotherapy, or targeted small molecule inhibitors. Semin Oncol. 2012;39(3):323–339. doi: 10.1053/j.seminoncol.2012.02.006.
    1. Drake CG. Combination immunotherapy approaches. Ann Oncol. 2012;23:viii41–viii46. doi: 10.1093/annonc/mds262.
    1. Torimura T, Iwamoto H, Nakamura T, Koga H, Ueno T, Kerbel RS, et al. Metronomic chemotherapy: possible clinical application in advanced hepatocellular carcinoma. Transl Oncol. 2013;6(5):511–519. doi: 10.1593/tlo.13481.
    1. Garcia-Aranda M, Redondo M. Immunotherapy: a challenge of breast cancer treatment. Cancers (Basel) 2019;11(12):1822. doi: 10.3390/cancers11121822.

Source: PubMed

3
Se inscrever