Safety and Activity of PolyPEPI1018 Combined with Maintenance Therapy in Metastatic Colorectal Cancer: an Open-Label, Multicenter, Phase Ib Study

Joleen M Hubbard, Enikő R Tőke, Roberto Moretto, Rondell P Graham, Hagop Youssoufian, Orsolya Lőrincz, Levente Molnár, Zsolt Csiszovszki, Jessica L Mitchell, Jaclynn Wessling, József Tóth, Chiara Cremolini, Joleen M Hubbard, Enikő R Tőke, Roberto Moretto, Rondell P Graham, Hagop Youssoufian, Orsolya Lőrincz, Levente Molnár, Zsolt Csiszovszki, Jessica L Mitchell, Jaclynn Wessling, József Tóth, Chiara Cremolini

Abstract

Purpose: Although chemotherapy is standard of care for metastatic colorectal cancer (mCRC), immunotherapy has no role in microsatellite stable (MSS) mCRC, a "cold" tumor. PolyPEPI1018 is an off-the-shelf, multi-peptide vaccine derived from 7 tumor-associated antigens (TAA) frequently expressed in mCRC. This study assessed PolyPEPI1018 combined with first-line maintenance therapy in patients with MSS mCRC.

Patients and methods: Eleven patients with MSS mCRC received PolyPEPI1018 and Montanide ISA51VG adjuvant subcutaneously, combined with fluoropyrimidine/biologic following first-line induction with chemotherapy and a biologic (NCT03391232). In Part A of the study, 5 patients received a single dose; in Part B, 6 patients received up to three doses of PolyPEPI1018 every 12 weeks. The primary objective was safety; secondary objectives were preliminary efficacy, immunogenicity at peripheral and tumor level, and immune correlates.

Results: PolyPEPI1018 vaccination was safe and well tolerated. No vaccine-related serious adverse event occurred. Eighty percent of patients had CD8+ T-cell responses against ≥3 TAAs. Increased density of tumor-infiltrating lymphocytes were detected post-treatment for 3 of 4 patients' liver biopsies, combined with increased expression of immune-related gene signatures. Three patients had objective response according to RECISTv1.1, and 2 patients qualified for curative surgery. Longer median progression-free survival for patients receiving multiple doses compared with a single dose (12.5 vs. 4.6 months; P = 0.017) suggested a dose-efficacy correlation. The host HLA genotype predicted multi-antigen-specific T-cell responses (P = 0.01) indicative of clinical outcome.

Conclusions: PolyPEPI1018 added to maintenance chemotherapy for patients with unresectable, MSS mCRC was safe and associated with specific immune responses and antitumor activity warranting further confirmation in a randomized, controlled setting.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Antitumor activity. A, Maximum changes from baseline in sum of target lesion size. B, Spider plot showing the changes of the target lesion sizes at each radiologic assessment (6-weekly). Each data point was compared with the lesion size measured at baseline. Dotted lines indicate patients receiving a single dose. CT scans and individual target lesion size changes for Patient 02–0004 (C), Patient 01–0004 (D), and Patient 01–0007 (E). Dotted lines denote post-study data for Patient 01–0007. Triangles indicate vaccination dates. Different gray shades on the line graphs and different colored arrows on the scans indicate different target lesions of the patients. *, This patient was considered with PD due to clinical progression by the investigator.
Figure 2.
Figure 2.
Durability of responses (post hoc analysis). A, Swimmer plot analysis for the individual disease courses prior-, during-, and post-study. B, Kaplan–Meier curve for the PFS of single- and multiple-dose groups. One patient (Patient 01–0008) from Part B who progressed after one dose and left the trial was included in the single dose group. Number of patients at risk is shown below the Kaplan–Meier curve. The Kaplan–Meier analysis contains censored data for curative surgery, RT, and LFU. PFS was calculated as the time from the date of initiating maintenance therapy to the date of first progression or censored. Post-study, the patients continued on maintenance treatment; for the 3 patients with curative surgery/RT, therapy was discontinued after surgery/RT. LFU, lost to follow-up; RT, radiotherapy.
Figure 3.
Figure 3.
Colorectal cancer–specific immune responses induced by PolyPEPI1018 at peripheral and tumor level indicate treatment benefit. A, The breadth of TAA-specific CD8+ T-cell responses and magnitude of CD4+ T-cell responses in each patient measured by IVS ELISpot. Post-vaccination (Po) results are the maximum responses measured for each subject during the study. CD8 T-cell responses were measured using the individual 9-mer test peptides listed in Supplementary Table S1, and summed for obtaining TAA-specific responses. CD4 T-cell responses were measured with the pooled 30-mer peptides. B, Number of responsive vaccine TAAs plotted by patient (n = 10). Dark gray bars: Pre-only: No change or <2-fold increase in responses compared with pre-vaccination; mid gray bars: response boosted compared with pre-vaccination (at least 2-fold increase by IVS ELISpot); light gray: de novo induced vaccine-specific immune responses (no pre-vaccination response measured). C, Magnitude of PolyPEPI1018 vaccine-specific CD8+ and CD4+ T-cell responses detected at baseline and after multiple doses. D, T-cell (TIL) infiltration to the CT and IM area of the tumors post-vaccination assessed by HalioDx's Immunoscore CR TL assay. Changes in TIL density were calculated from IHC data obtained for pre/post-vaccination biopsy pairs; pre: baseline sample (except for Patient 01–0002, it was week 12 sample); Post: tumor biopsy at week 12 (for Patients 02–0002 and 02–0004) or week 38 (Patients 01–0002 and 01–0007). E, Whisker plot of upregulated gene-expression signatures upon treatment with PolyPEPI1018 for responder (R) tumors (Patients 02–0004, 01–0002, and 01–0007) and a nonresponder (NR) tumor (Patient 02–0002) assessed by NanoString's PanCancer gene-expression panel. Each dot represents the fold change of a gene in a sample. Boxes on the plot represents the quartiles (1 to 3). CD8+ effector T-cell gene signature: [CD8A molecule (CD8A), CD8B molecule (CD8B), eomesodermin (EOMES), granzyme A (GZMA), granzyme B (GZMB), IFNγ (IFNG), and perforin 1 (PRF1)]; IFNγ gene signature: [indoleamine 2,3-dioxygenase 1 (IDO1), C-X-C motif chemokine ligand (CXCL) 9 and 10, MHC class II DR α (HLA-DRA), signal transducer and activator of transcription 1 (STAT1) and IFNG] and PD-L1 and PD-L2 gene signatures. F, Impact of TAA-specific CD8+ T-cell responses boosted by the vaccine (measured by IVS ELISpot) on treatment benefit; DCB, durable clinical benefit = patients with objective tumor response (PR) and/or stabilized disease (SD) for at least 50 weeks on maintenance treatment; Others: patients with no DCB. ΔSFU, background corrected spot-forming units; #, number. ND, not detected; ns., not significant. Error bars represent SEM.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69:7–34.
    1. Saunders M, Iveson T. Management of advanced colorectal cancer: state of the art. Br J Cancer 2006;95:131–8.
    1. Cremolini C, Antoniotti C, Rossini D, Lonardi S, Loupakis F, Pietrantonio F, et al. . Upfront FOLFOXIRI plus bevacizumab and reintroduction after progression versus mFOLFOX6 plus bevacizumab followed by FOLFIRI plus bevacizumab in the treatment of patients with metastatic colorectal cancer (TRIBE2): a multicenter, open-label, phase III, randomized, controlled trial. Lancet Oncol 2020;21:497–507.
    1. Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, et al. . Effect of first-line chemotherapy combined with cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal cancer: a randomized clinical trial. JAMA 2017;317:2392–401.
    1. Moretto R, Cremolini C, Rossini D, Pietrantonio F, Battaglin F, Mennitto A, et al. . Location of primary tumor and benefit from anti-epidermal growth factor receptor monoclonal antibodies in patients with RAS and BRAF wild-type metastatic colorectal cancer. Oncologist 2016;21:988–94.
    1. Tejpar S, Stintzing S, Ciardiello F, Tabernero J, Van Cutsem E, Beier F, et al. . Prognostic and predictive relevance of primary tumor location in patients with RAS wild-type metastatic colorectal cancer: retrospective analyses of the CRYSTAL and FIRE-3 trials. JAMA Oncol 2017;3:194–201.
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumors with combination immunotherapies. Nat Rev Drug Discov 2019;18:197–218.
    1. Boisen MK, Johansen JS, Dehlendorff C, Larsen JS, Østerlind K, Hansen J, et al. . Primary tumor location and bevacizumab effectiveness in patients with metastatic colorectal cancer. Ann Oncol 2013;24:2554–9.
    1. Tournigand C, Cervantes A, Figer A, Lledo G, Flesch M, Buyse M, et al. . OPTIMOX1: a randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in a stop-and-go fashion in advanced colorectal cancer–a GERCOR study. J Clin Oncol 2006;24:394–400.
    1. Hegewisch-Becker S, Graeven U, Lerchenmuller CA, Killing B, Depenbusch R, Steffens CC, et al. . Maintenance strategies after first-line oxaliplatin plus fluoropyrimidine plus bevacizumab for patients with metastatic colorectal cancer (AIO 0207): a randomized, noninferiority, open-label, phase III trial.Lancet Oncol 2015;16:1355–69.
    1. Hubbard JM, Grothey A. When less is more: maintenance therapy in colorectal cancer. Lancet 2015;385:1808–10.
    1. Koeberle D, Betticher DC, von Moos R, Dietrich D, Brauchli P, Baertschi D, et al. . Bevacizumab continuation versus no continuation after first-line chemotherapy plus bevacizumab in patients with metastatic colorectal cancer: a randomized phase III noninferiority trial (SAKK 41/06). Ann Oncol 2015;26:709–14.
    1. Simkens LH, van Tinteren H, May A, ten Tije AJ, Creemers GJ, Loosveld OJ, et al. . Maintenance treatment with capecitabine and bevacizumab in metastatic colorectal cancer (CAIRO3): a phase III randomized controlled trial of the Dutch colorectal cancer group. Lancet 2015;385:1843–52.
    1. Tournigand C, Chibaudel B, Samson B, Scheithauer W, Vernerey D, Mésange P, et al. . Bevacizumab with or without erlotinib as maintenance therapy in patients with metastatic colorectal cancer (GERCOR DREAM; OPTIMOX3): a randomized, open-label, phase III trial. Lancet Oncol 2015;16:1493–505.
    1. Kreidieh M, Mukherji D, Temraz S, Shamseddine A. Expanding the scope of immunotherapy in colorectal cancer: current clinical approaches and future directions. Biomed Res Int 2020;2020:9037217.
    1. Grothey A, Tabernero J, Arnold D, De Gramont A, Ducreux MP, O'Dwyer PJ, et al. . Fluoropyrimidine (FP) + bevacizumab (BEV) + atezolizumab vs FP/BEV in BRAFwt metastatic colorectal cancer (mCRC): findings from cohort 2 of MODUL – a multicenter, randomized trial of biomarker-driven maintenance treatment following first-line induction therapy. Ann Oncol 2018;29:viii714–5.
    1. Mansfield AS, Nevala WK, Lieser EA, Leontovich AA, Markovic SN. The immunomodulatory effects of bevacizumab on systemic immunity in patients with metastatic melanoma. Oncoimmunology 2013;2:e24436.
    1. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res 2015;3:436–43.
    1. Galluzzi L, Humeau J, Buqué A, Zitvogel L, Kroemer G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol 2020;17:725–41.
    1. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunologic effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 2015;28:690–714.
    1. Gmeiner WH. Fluoropyrimidine modulation of the antitumor immune response-prospects for improved colorectal cancer treatment. Cancers 2020;12:1641.
    1. Fang H, Ang B, Xu X, Huang X, Wu Y, Sun Y, et al. . TLR4 is essential for dendritic cell activation and antitumor T-cell response enhancement by DAMPs released from chemically stressed cancer cells. Cell Mol Immunol 2014;11:150–9.
    1. Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. . 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell–dependent antitumor immunity. Cancer Res 2010;70:3052–61.
    1. Roland CL, Lynn KD, Toombs JE, Dineen SP, Udugamasooriya DG, Brekken RA. Cytokine levels correlate with immune cell infiltration after anti-VEGF therapy in preclinical mouse models of breast cancer. PLoS One 2009;4:e7669.
    1. Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet A-L, et al. . VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 2015;212:139–48.
    1. Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. . Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun 2016;7:12624.
    1. Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, et al. . Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 2011;91:1071–121.
    1. Zheng L, Xie G, Duan G, Yan X, Li Q. High expression of testes-specific protease 50 is associated with poor prognosis in colorectal carcinoma. PLoS One 2011;6:e22203.
    1. Goossens-Beumer IJ, Zeestraten EC, Benard A, Christen T, Reimers MS, Keijzer R, et al. . Clinical prognostic value of combined analysis of Aldh1, Survivin, and EpCAM expression in colorectal cancer. Br J Cancer 2014;110:2935–44.
    1. Choi J, Chang H. The expression of MAGE and SSX, and correlation of COX2, VEGF, and survivin in colorectal cancer. Anticancer Res 2012;32:559–64.
    1. Kanojia D, Garg M, Gupta S, Gupta A, Suri A. Sperm-associated antigen 9 is a novel biomarker for colorectal cancer and is involved in tumor growth and tumorigenicity. Am J Pathol 2011;178:1009–20.
    1. Dakshinamurthy AG, Ramesar R, Goldberg P, Blackburn JM. Infrequent and low expression of cancer-testis antigens located on the X chromosome in colorectal cancer: implications for immunotherapy in South African populations. Biotechnol J 2008;3:1417–23.
    1. Song MH, Ha JC, Lee SM, Park YM, Lee SY. Identification of BCP-20 (FBXO39) as a cancer/testis antigen from colon cancer patients by SEREX. Biochem Biophys Res Commun 2011;408:195–201.
    1. Hasegawa H, Mori M, Haraguchi M, Ueo H, Sugimachi K, Akiyoshi T. Expression spectrum of melanoma antigen-encoding gene family members in colorectal carcinoma. Arch Pathol Lab Med 1998;122:551–4.
    1. Hubbard JM, Cremolini C, Graham RP, Moretto R, Mitchell JL, Wessling J, et al. . A phase I study of PolyPEPI1018 vaccine plus maintenance therapy in patients with metastatic colorectal cancer with a predictive biomarker (OBERTO). J Clin Oncol 2019;37:3557.
    1. Somogyi E, Csiszovszki Z, Molnar L, Lorincz O, Toth J, Pattijn S, et al. . A peptide vaccine candidate tailored to individuals' genetics mimics the multi-targeted T-cell immunity of COVID-19 convalescent subjects. Front Genet 2021;12:684152.
    1. Lorincz O, Toth J, Molnar L, Miklos I, Pantya K, Megyesi M, et al. . In silico model estimates the clinical trial outcome of cancer vaccines. Cells 2021;10:3048.
    1. Saxena M, van der Burg SH, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer 2021;21:360–78.
    1. Wang Y, Lin HC, Huang MY, Shao Q, Wang ZQ, Wang FH, et al. . The Immunoscore system predicts prognosis after liver metastasectomy in colorectal cancer liver metastases. Cancer Immunol Immunother 2018;67:435–44.
    1. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. . IFNγ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40.
    1. van Doorn E, Liu H, Huckriede A, Hak E. Safety and tolerability evaluation of the use of Montanide ISA51 as vaccine adjuvant: a systematic review. Hum Vaccin Immunother 2016;12:159–69.
    1. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. . Proteomics. Tissue-based map of the human proteome. Science 2015;23;347:1260419.
    1. Yi F, Ni W, Liu W, Pan X, Han X, Yang L, et al. . SPAG9 is overexpressed in human astrocytoma and promotes cell proliferation and invasion. Tumour Biol 2013;34:2849–55.
    1. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbé C, et al. . Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 2009;15:7412–20.
    1. Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. . Multi-peptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012;18:1254–61.
    1. Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. . An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017;547:217–21.
    1. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Lower M, et al. . Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017;547:222–6.
    1. Toke ER, Megyesi M, Molnar L, Tóth J, Lőrincz O, van der Burg SH, et al. . Prediction the clinical outcomes of cancer patients after peptide vaccination. J Clin Oncol 2019;37(15_suppl):e14295.
    1. Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, et al. . Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018;359:582–7.

Source: PubMed

3
Tilaa