Implication of combined PD-L1/PD-1 blockade with cytokine-induced killer cells as a synergistic immunotherapy for gastrointestinal cancer

Congqi Dai, Fengjuan Lin, Ruixuan Geng, Xiaoxiao Ge, Wenbo Tang, Jinjia Chang, Zheng Wu, Xinyang Liu, Ying Lin, Zhe Zhang, Jin Li, Congqi Dai, Fengjuan Lin, Ruixuan Geng, Xiaoxiao Ge, Wenbo Tang, Jinjia Chang, Zheng Wu, Xinyang Liu, Ying Lin, Zhe Zhang, Jin Li

Abstract

Cytokine-induced killer (CIK) cells represent a realistic approach in cancer immunotherapy with confirmed survival benefits in the context of metastatic solid tumors. However, therapeutic effects are limited to a fraction of patients. In this study, immune-resistance elements and ideal combination therapies were explored. Initially, phenotypic analysis was performed to document CD3, CD56, NKG2D, DNAM-1, PD-L1, PD-1, CTLA-4, TIM-3, 2B4, and LAG-3 on CIK cells. Upon engagement of CIK cells with the tumor cells, expression of PD-1 on CIK cells and PD-L1 on both cells were up-regulated. Over-expression of PD-L1 levels on tumor cells via lentiviral transduction inhibited tumoricidal activity of CIK cells, and neutralizing of PD-L1/PD-1 signaling axis could enhance their tumor-killing effect. Conversely, blockade of NKG2D, a major activating receptor of CIK cells, largely caused dysfunction of CIK cells. Functional study showed an increase of NKG2D levels along with PD-L1/PD-1 blockade in the presence of other immune effector molecule secretion. Additionally, combined therapy of CIK infusion and PD-L1/PD-1 blockade caused a delay of in vivo tumor growth and exhibited a survival advantage over untreated mice. These results provide a preclinical proof-of-concept for simultaneous PD-L1/PD-1 pathways blockade along with CIK infusion as a novel immunotherapy for unresectable cancers.

Keywords: CIK; NKG2D; PD-L1/PD-1; gastrointestinal tumor; immunotherapy.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors disclose no potential conflicts of interest

Figures

Figure 1. In vitro expansion and main…
Figure 1. In vitro expansion and main phenotypes of CIK cells derived from donors (n = 10)
Differential expression of three main phenotypic subsets of CIK cells over in vitro culture are shown for identification of CIK cells, CD3/CD56 (A), T cell associated immune receptors, CD3/CD4/CD8 (B), and major activating receptors of CIK cells, CD3/NKG2D (C). Flow cytometric analysis was performed over expansion periods on day0, day7, day14, and day21. Each representative picture was accordingly shown on the right panel.
Figure 2. Stable variations in PD-L1 expression…
Figure 2. Stable variations in PD-L1 expression on the tumor cells via lentiviral transduction and corresponding influences upon tumoricidal activity of CIK cells
A. Gastric and colorectal cancer cell lines (MGC803 and RKO) were transduced with lentiviral vectors containing siRNA directed against PD-L1, whereas HGC27 and SW480 were transfected with PD-L1 cDNA. After 72 hours, transfected tumor cells were confirmed for PD-L1 expression by flow cytometry. filled isotype staining; bold line PD-L1-stained untransfected cell lines; dashed line PD-L1-stained transfected cell lines with siRNA (MGC803 and RKO) or PD-L1 cDNA (HGC27 and SW480). B. CIK cells exerting cytotoxicity against the tumor cells were assessed by a non-radioactive cytotoxicity assay. Comparative curves were drawn between the tumor cells transfected with siRNA or PD-L1 cDNA and with scrambled shRNA as negative control group. Results represent at least three independent experiments using CIK cells from the same donor and are shown as Mean±SEM. C. CIK cells was pre-incubated with above tumor cells in 2ml culture medium per well in a 6-well-plate at a E:T ratio of 10:1 overnight, and both suspending and adherent cells were harvested and analyzed for cell apoptosis assay. D. Concordantly, obvious lysis of representative tumor cells (MGC803 and RKO) was observed over time by microscopy.
Figure 3. PD-L1/PD-1 pathway blockade efficiently increases…
Figure 3. PD-L1/PD-1 pathway blockade efficiently increases tumor-killing activity of CIK cells
A. The levels of PD-1 and PD-L1 on the CIK cells were observed after 12 hours co-culture with MGC803 or RKO. Data from three independent experiments were shown as Mean±SEM, and statistical significance was determined by Student t test (* means P<0.05, ** means P<0.01, and *** means P<0.001). Representative pictures are indicated on the right panel. B. PD-L1 levels on the tumor cells (MGC803) were detected by the co-culture, as well as those with the adding of IFN-γ (200U/ml). filled isotype staining; bold line PD-L1-stained MGC803. C. Correlation was performed between IFN-γ and PD-L1 mRNA levels in the gastric cancer tissues. Pearson's r and P value are displayed. -ΔCt indicates the difference in the threshold cycle between the target genes and β-actin. D. CIK cells exerting cytotoxicity against MGC803 or RKO, respectively, were assessed by a non-radioactive cytotoxicity assay, with or without PD-L1/PD-1 blockade using indicated antibodies (20μg/ml). Corresponding isotype antibody (20μg/ml) for PD-L1/PD-1 was used as a control group. Results represent at least three independent experiments using the CIK cells from the same donor and are shown as Mean±SEM.
Figure 4. In vivo cytotoxicity of CIK…
Figure 4. In vivo cytotoxicity of CIK cells against murine gastric cancer model
Nude mice were subcutaneously implanted with an 8mm3 tumor fragment prepared beforehand with MGC803 cells injected into 5 mice. Three days after tumor implantation, mice received weekly intravenous infusions of 107 CIK cells. Anti-PD-1 antibody was pre-added to block PD-1 on CIK cells for 1 hour. A. An obvious tumor growth delay was observed in combined CIK with anti-PD-1-treated mice compared with the control group (P<0.001), whereas no significant difference was found in CIK alone-treated mice (P>0.05). B. Percentage survival of MGC803 tumor-bearing mice treated with CIK cells with or without anti-PD-1 antibody was significantly increased compared with untreated mice using Kaplan-Meier analysis (P<0.05), whereas no significant difference was found between CIK cells-treated groups with or without anti-PD-1 antibody (P>0.05). C. At the end of infusion, representative pictures of CIK cells infiltration at tumor sites were shown by IHC using antibodies against CD5, and expression of ki-67 was shown in tumors from CIK-treated mice and control, scale bar, 50μm. Tumor growth and survival data were shown independently with 6 to 10 mice per group.
Figure 5. Blocking of NKG2D binding with…
Figure 5. Blocking of NKG2D binding with its ligands results in impaired cytotoxic activity of CIK cells against tumor cell lines (MGC803, HGC27, RKO, and SW480)
A. All the tumor cell lines were observed for MIC A/B expression. Besides, the lytic activity of CIK cells, with or without blocking its recognition receptor, NKG2D, was assessed by cytotoxicity assay. B. Meanwhile, all the above cell lines were detected for the levels of ULBP-2, another crucial ligand for NKG2D. Antibody-blockade of ULBP-2 was conducted to observe cytotoxicity of CIK cells against all the above cells that differentially express ULBP-2. filled isotype staining; bold line MIC A/B− or ULBP-2-stained tumor cell. All the data on the curves represent three independent experiments using CIK cells from the same donor and are shown as Mean±SEM.
Figure 6. Disrupting PD-L1/PD-1 binding using blocking…
Figure 6. Disrupting PD-L1/PD-1 binding using blocking antibodies markedly induced an increase in multiple immune effector molecules
A. IFN-γ and CD107a, as direct measurements of cytotoxicity of CIK cells against tumor cells, as well as granzyme B, were observed by the co-incubation at E:T ratio of 5:1, along with the detection for a potentially responsive phenotype in terms of the co-inhibitory molecules, CTLA-4 and LAG-3 (B), and the activating receptors, NKG2D and DNAM-1 (C). D. Gastric clinical specimens were examined for analyzing the difference in the mRNA levels of these functional receptors between the gastric cancer samples and matched adjacent normal samples, and statistical significance was determined by paired t test. E. Associated immune-promoting molecules, IFN-γ and CD107a, accompanied with NKG2D levels, were examined after PD-L1/PD-1 pathway blockade using indicated antibodies (20μg/ml), among the test groups and the control groups (indicated as untreated group and isotype antibody group). F. 5×105 CIK cells were co-cultured in triplicate with 1×105 tumor cells in 500 μl culture medium per well in a 48-well-plate. Anti-PD-L1 and anti-PD-1 together with isotype antibody (20μg/ml) were separately added into each well. After 48 h, the levels of IFN-γ in the supernatants were evaluated by ELISA. G. Using the same co-culture condition described above, changes of NKG2D levels were observed with the direct adding of IFN-γ cytokine. Results represent three independent experiments and are shown as Mean±SEM (* means P<0.05, ** means P<0.01, and *** means P<0.001).

References

    1. Power DG, Kelsen DP, Shah MA. Advanced gastric cancer--slow but steady progress. Cancer Treat Rev. 2010;36:384–92.
    1. Rosenberg SA. Cell transfer immunotherapy for metastatic solid cancer--what clinicians need to know. Nat Rev Clin Oncol. 2011;8:577–85.
    1. Drake CG. Combination immunotherapy approaches. Ann Oncol. 2012;23:i41–6.
    1. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986;233:1318–21.
    1. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, Matory YL, Skibber JM, Shiloni E, Vetto JT, Seipp CA, Simpson C, Reichert CM. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med. 1985;313:1485–92.
    1. Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med. 1991;174:139–49.
    1. Baker J, Verneris MR, Ito M, Shizuru JA, Negrin RS. Expansion of cytolytic CD8 (+) natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon gamma production. Blood. 2001;97:2923–31.
    1. Hongeng S, Petvises S, Worapongpaiboon S, Rerkamnuaychoke B, Pakakasama S, Jootar S. Generation of CD3+ CD56+ cytokine-induced killer cells and their in vitro cytotoxicity against pediatric cancer cells. Int J Hematol. 2003;77:175–9.
    1. Kim HM, Lim J, Yoon YD, Ahn JM, Kang JS, Lee K, Park SK, Jeong YJ, Kim JM, Han G, Yang KH, Kim YJ, Kim Y, et al. Anti-tumor activity of ex vivo expanded cytokine-induced killer cells against human hepatocellular carcinoma. Int Immunopharmacol. 2007;7:1793–801.
    1. Kuçi S, Rettinger E, Voss B, Weber G, Stais M, Kreyenberg H, Willasch A, Kuçi Z, Koscielniak E, Klöss S, von Laer D, Klingebiel T, Bader P. Efficient lysis of rhabdomyosarcoma cells by cytokine-induced killer cells: implications for adoptive immunotherapy after allogeneic stem cell transplantation. Haematologica. 2010;95:1579–86.
    1. Jamieson AM, Diefenbach A, McMahon CW, Xiong N, Carlyle JR, Raulet DH. The role of the NKG2D immunoreceptor in immune cell activation and natural killing. Immunity. 2002;17:19–29.
    1. Mehta BA, Schmidt-Wolf IG, Weissman IL, Negrin RS. Two pathways of exocytosis of cytoplasmic granule contents and target cell killing by cytokine-induced CD3+ CD56+ killer cells. Blood. 1995;86:3493–9.
    1. Pende D, Rivera P, Marcenaro S, Chang CC, Biassoni R, Conte R, Kubin M, Cosman D, Ferrone S, Moretta L, Moretta A. Major histocompatibility complex class I-related chain A and UL16-binding protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res. 2002;62:6178–86.
    1. Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci U S A. 1996;93:12445–50.
    1. Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T. Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc Natl Acad Sci U S A. 1999;96:6879–84.
    1. Liu L, Zhang W, Qi X, Li H, Yu J, Wei S, Hao X, Ren X. Randomized study of autologous cytokine-induced killer cell immunotherapy in metastatic renal carcinoma. Clin Cancer Res. 2012;18:1751–9.
    1. Introna M, Borleri G, Conti E, Franceschetti M, Barbui AM, Broady R, Dander E, Gaipa G, D'Amico G, Biagi E, Parma M, Pogliani EM, Spinelli O. Repeated infusions of donor-derived cytokine-induced killer cells in patients relapsing after allogeneic stem cell transplantation: a phase I study. Haematologica. 2007;92:952–9.
    1. Olioso P, Giancola R, Di Riti M, Contento A, Accorsi P, Iacone A. Immunotherapy with cytokine induced killer cells in solid and hematopoietic tumours: a pilot clinical trial. Hematol Oncol. 2009;27:130–9.
    1. Takayama T, Sekine T, Makuuchi M, Yamasaki S, Kosuge T, Yamamoto J, Shimada K, Sakamoto M, Hirohashi S, Ohashi Y, Kakizoe T. Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet. 2000;356:802–7.
    1. Hontscha C, Borck Y, Zhou H, Messmer D, Schmidt-Wolf IG. Clinical trials on CIK cells: first report of the international registry on CIK cells (IRCC) J Cancer Res Clin Oncol. 2011;137:305–10.
    1. Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765–72.
    1. Moll M, Kuylenstierna C, Gonzalez VD, Andersson SK, Bosnjak L, Sönnerborg A, Quigley MF, Sandberg JK. Severe functional impairment and elevated PD-1 expression in CD1d-restricted NKT cells retained during chronic HIV-1 infection. Eur J Immunol. 2009;39:902–11.
    1. Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206:1327–37.
    1. Nomi T, Sho M, Akahori T, Hamada K, Kubo A, Kanehiro H, Nakamura S, Enomoto K, Yagita H, Azuma M, Nakajima Y. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res. 2007;13:2151–7.
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–44.
    1. Thompson RH, Dong H, Kwon ED. Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clin Cancer Res. 2007;13:709–15.
    1. Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin Cancer Res. 2004;10:5094–100.
    1. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99:12293–7.
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800.
    1. Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, Gajewski TF. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64:1140–5.
    1. Blank C, Kuball J, Voelkl S, Wiendl H, Becker B, Walter B, Majdic O, Gajewski TF, Theobald M, Andreesen R, Mackensen A. Blockade of PD-L1 (B7-H1) augments human tumor-specific T cell responses in vitro. Int J Cancer. 2006;119:317–27.
    1. Strome SE, Dong H, Tamura H, Voss SG, Flies DB, Tamada K, Salomao D, Cheville J, Hirano F, Lin W, Kasperbauer JL, Ballman KV, Chen L. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 2003;63:6501–5.
    1. Sangiolo D, Mesiano G, Gammaitoni L, Leuci V, Todorovic M, Giraudo L, Cammarata C, Dell'Aglio C, D'Ambrosio L, Pisacane A, Sarotto I, Miano S, Ferrero I, et al. Cytokine-induced killer cells eradicate bone and soft-tissue sarcomas. Cancer Res. 2014;74:119–29.
    1. Zhang Q, Liu XY, Zhang T, Zhang XF, Zhao L, Long F, Liu ZK, Wang EH. The dual-functional capability of cytokine-induced killer cells and application in tumor immunology. Hum Immunol. 2015;76:385–91.
    1. Mesiano G, Todorovic M, Gammaitoni L, Leuci V, Giraudo Diego L, Carnevale-Schianca F, Fagioli F, Piacibello W, Aglietta M, Sangiolo D. Cytokine-induced killer (CIK) cells as feasible and effective adoptive immunotherapy for the treatment of solid tumors. Expert Opin Biol Ther. 2012;12:673–84.
    1. Blackburn SD, Crawford A, Shin H, Polley A, Freeman GJ, Wherry EJ. Tissue-specific differences in PD-1 and PD-L1 expression during chronic viral infection: implications for CD8 T-cell exhaustion. J Virol. 2010;84:2078–89.
    1. Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, Bettini ML, Gravano DM, Vogel P, Liu CL, Tangsombatvisit S, Grosso JF, Netto G, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72:917–27.
    1. Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116:1935–45.
    1. Shafi S, Vantourout P, Wallace G, Antoun A, Vaughan R, Stanford M, Hayday A. An NKG2D-mediated human lymphoid stress surveillance response with high interindividual variation. Sci Transl Med. 2011;3:113–124.
    1. Diefenbach A, Jensen ER, Jamieson AM, Raulet DH. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature. 2001;413:165–71.
    1. McGilvray RW, Eagle RA, Rolland P, Jafferji I, Trowsdale J, Durrant LG. ULBP2 and RAET1E NKG2D ligands are independent predictors of poor prognosis in ovarian cancer patients. Int J Cancer. 2010;127:1412–20.
    1. de Kruijf EM, Sajet A, van Nes JG, Putter H, Smit VT, Eagle RA, Jafferji I, Trowsdale J, Liefers GJ, van de Velde CJ, Kuppen PJ. NKG2D ligand tumor expression and association with clinical outcome in early breast cancer patients: an observational study. Bmc Cancer. 2012;12:24.
    1. Osaki T, Saito H, Yoshikawa T, Matsumoto S, Tatebe S, Tsujitani S, Ikeguchi M. Decreased NKG2D expression on CD8+ T cell is involved in immune evasion in patients with gastric cancer. Clin Cancer Res. 2007;13:382–387.
    1. Saito H, Osaki T, Ikeguchi M. Decreased NKG2D expression on NK cells correlates with impaired NK cell function in patients with gastric cancer. Gastric Cancer. 2012;15:27–33.
    1. Marcais A, Viel S, Grau M, Henry T, Marvel J, Walzer T. Regulation of mouse NK cell development and function by cytokines. Front Immunol. 2013;4:450.
    1. Nishimura R, Baker J, Beilhack A, Zeiser R, Olson JA, Sega EI, Karimi M, Negrin RS. In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood. 2008;112:2563–74.

Source: PubMed

3
Abonneren