Inhibiting Immune Checkpoints for the Treatment of Bladder Cancer

S Bidnur, R Savdie, P C Black, S Bidnur, R Savdie, P C Black

Abstract

Background: Increasing evidence supporting the role of immune checkpoint blockade in cancer management has been bolstered by recent reports demonstrating significant and durable clinical responses across multiple tumour types, including metastatic urothelial carcinoma (mUC). The majority of these results are achieved via blockade of the programmed death (PD) axis, which like CTLA-4 blockade permits T-cell activation and immune-mediated anti-tumour activity- essentially harnessing the patient's own immune system to mount an anti-neoplastic response. However, while clinical responses can be striking, our understanding of the biology of immune checkpoint blockade is only beginning to shed light on how to maximize and even improve patient outcomes with immune checkpoint blockade, especially in UC.

Methods: We performed a literature review for immune checkpoint blockade with a focus on rationale for checkpoint therapy and outcomes in UC. We also highlight the advances made in other tumour types, with a focus on the recent 2015 meeting of the American Society for Clinical Oncology.

Results: In heavily pre-treated UC, trials are suggesting objective response rates above 30% . These impressive results are seen across multiple different tumour types, especially those with high burden of DNA level mutations. Identification of prognostic biomarkers is currently under investigation, in order to improve patient selection. Interestingly, response to PD-1 directed therapy is seen even in patients with no evidence of PD-1 positivity on immunohistochemistry. This has led to the development of enhanced biomarkers including assessing DNA mutation rates and immune gene signatures, to improve patient selection.

Conclusions: Immune checkpoint blockade is an exciting cancer treatment modality which is demonstrating impressive clinical results across multiple tumour types. For UC, anti-PD directed therapy represents a much needed treatment in the metastatic, post chemotherapy context. Potential for these agents to have clinical utility in non-metastatic UC is still to be assessed.

Keywords: Bladder cancer; PD-1/PD-L1; immune checkpoint blockade; predictive biomarkers.

References

    1. Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–723.
    1. Robert C, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011;364:2517–2526.
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–2465.
    1. Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–2454.
    1. Tomihara K, Curiel TJ, Zhang B. Optimization of immunotherapy in elderly cancer patients. Crit Rev Oncog 2013;18:573–583.
    1. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011;331:1565–1570.
    1. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 2015;160:48–61.
    1. Dong H, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat Med 2002;8:793–800.
    1. Chen DS, Mellman I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013;39:1–10.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell 2011;144:646–674.
    1. Pardoll DM. T he blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–264.
    1. Carosella ED, Ploussard G, LeMaoult J, Desgrandchamps F. A Systematic Review of Immunotherapy in Urologic Cancer: Evolving Roles for Targeting of CTLA-4, PD-1/PD-L1, and HLA-G. Eur Urol 2015;68:267–279.
    1. Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 SYSTEM OF T CELL COSTIMULATION. Annual Review of Immunology 1996;14:233–258.
    1. Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev 2009;229:12–26.
    1. Tivol EA, et al. CTLA4Ig prevents lymphoproliferation and fatal multiorgan tissue destruction in CTLA-4-deficient mice. J Immunol 1997;158:5091–5094.
    1. Ernst B, Anderson KS. Immunotherapy for the treatment of breast cancer. Curr Oncol Rep 2015;17:5.
    1. Simeone E, Ascierto PA. Immunomodulating antibodies in the treatment of metastatic melanoma: The experience with anti-CTLA-4, anti-CD137, and anti-PD1. J Immunotoxicol 2012;9:241–247.
    1. Freeman GJ, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027–1034.
    1. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med 2015;21:24–33.
    1. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11:3887–3895.
    1. Francisco LM, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 2009;206:3015–3029.
    1. Tsushima F, et al. Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood 2007;110:180–185.
    1. Fanoni D, et al. New monoclonal antibodies against B-cell antigens: Possible new strategies for diagnosis of primary cutaneous B-cell lymphomas. Immunol Lett 2011;134:157–160.
    1. Ahmadzadeh M, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114:1537–1544.
    1. Wilke CM, et al. Dual biological effects of the cytokines interleukin-10 and interferon-gamma. Cancer Immunol Immunother 2011;60:1529–1541.
    1. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011;480:480–489.
    1. Morales A, Eidinger D, Bruce AW. Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J Urol 1976;116:180–183.
    1. De Boer EC, et al. Presence of activated lymphocytes in the urine of patients with superficial bladder cancer after intravesical immunotherapy with bacillus Calmette-Guerin. Cancer Immunol Immunother 1991;33:411–416.
    1. Eto M, et al. Importance of urinary interleukin-18 in intravesical immunotherapy with bacillus calmette-guerin for superficial bladder tumors. Urol Int 2005;75:114–118.
    1. Jackson AM, et al. Changes in urinary cytokines and soluble intercellular adhesion molecule-1 (ICAM-1) in bladder cancer patients after bacillus Calmette-Guerin (BCG) immunotherapy. Clin Exp Immunol 1995;99:369–375.
    1. Morton D, Eilber FR, Malmgren RA, Wood WC. Immunological factors which influence response to immunotherapy in malignant melanoma. Surgery 1970;68:158–163; discussion 163–4.
    1. Ratliff TL, Ritchey JK, Yuan JJ, Andriole GL, Catalona WJ. T-cell subsets required for intravesical BCG immunotherapy for bladder cancer. J Urol 1993;150:1018–1023.
    1. Redelman-Sidi G, Glickman MS, Bochner BH. The mechanism of action of BCG therapy for bladder cancer–a current perspective. Nat Rev Urol 2014;11:153–162.
    1. Inman BA, et al. PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: Associations with localized stage progression. Cancer 2007;109:1499–1505.
    1. Nakanishi J, et al. Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother 2007;56:1173–1182.
    1. Boorjian SA, et al. T-cell coregulatory molecule expression in urothelial cell carcinoma: Clinicopathologic correlations and association with survival. Clin Cancer Res 2008;14:4800–4808.
    1. Xylinas E, et al. Association of T-cell co-regulatory protein expression with clinical outcomes following radical cystectomy for urothelial carcinoma of the bladder. Eur J Surg Oncol 2014;40:121–127.
    1. Bellmunt J, et al. Association of PD-L1 expression on tumor-infiltrating mononuclear cells and overall survival in patients with urothelial carcinoma. Ann Oncol 2015;26:812–817.
    1. Kandoth C, et al. Mutational landscape and significance across 12 major cancer types. Nature 2013;502:333–509.
    1. Cancer Genome Atlas Research N. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014;507:315–322.
    1. Rizvi NA, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science(2015).
    1. Cazier JB, et al. Whole-genome sequencing of bladder cancers reveals somatic CDKN1A mutations and clinicopathological associations with mutation burden. Nat Commun 2014;5:3756.
    1. Hurwitz MA, Yao A, Hafez X, Schalper N, Rimm K, Petrylak DD. The effect of BCG intravesical therapy and recurrence on PDL1 expression in non-invasive bladder cancers. J Clin Oncol 2015;33:e15504.
    1. Baras AAM, Guner M, Gandhi G, Liu N, Taube J, Bivalacqua J, Netto TG. PDL1 status in muscle invasive urothelial bladder carcinoma (MIBC) in the context of neoadjuvant cisplatin based chemotherapy. J Urol 2015;193:e703.
    1. Choi W, et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 2014;25:152–165.
    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.
    1. Powles T, et al. MPDLA (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 2014;515:558–562.
    1. Petrylak D, et al. A phase Ia study of MPDLA (anti-PDL1): Updated response and survival data in urothelial bladder cancer (UBC). J Clin Oncol 2015;33(15_suppl), 4501.
    1. Plimack E, et al. Pembrolizumab (MK-3475) for advanced urothelial cancer: Updated results and biomarker analysis from KEYNOTE-012. J Clin Oncol 2015;33(suppl); abstr4502.
    1. Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDLA in cancer patients. Nature 2014;515:563–567.
    1. Buisseret L, Specht J, Dees EC, Berger R, Gupta S, Geva R, Pusztai L, Gause CK, Karantza V, Nanda R. KEYNOTE- A phase Ib study of pembrolizumab (MK-in patients (pts) with metastatic triple-negative breast cancer (mTNBC). Ann Oncol 2015;26:6.
    1. Gettinger S, et al. Molecular, immune and histopathological characterization of NSCLC based on PDL1 expression on tumor and immune cells and association with response to the anti-PDL1 antibody MPDLA3280A. J Clin Oncol 2015;33(suppl); abstr 3015.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69–74.
    1. Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–128.
    1. Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–2199.
    1. Rajasagi M, et al. Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood 2014;124:453–462.
    1. Le DT, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509–2520.
    1. Weber JS, Kahler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol 2012;30:2691–2697.
    1. Postow MA, et al. Nivolumab and Ipilimumab versus Ipilimumab in Untreated Melanoma. New England Journal of Medicine 2015;372:2006–2017.
    1. Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–422.
    1. Nakamoto N, et al. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathog 2009;5:e1000313.
    1. Larkin J, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015;373:23–34.
    1. Spigel D, et al. Clinical activity and safety from a phase II study (FIR) of MPDLA (anti-PDL1) in PD-L1–selected patients with non-small cell lung cancer (NSCLC). J Clin Oncol 2015;33(suppl); abstr 8028.
    1. Spira A, et al. Efficacy, safety and predictive biomarker results from a randomized phase II study comparing MPDLA vs docetaxel in 2L/3L NSCLC (POPLAR). J Clin Oncol 2015;33(suppl); abstr 8010.
    1. Damrauer JS, et al. 2014; Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc Natl Acad Sci U S A 2015;111:3110–3115.
    1. Gatalica Z, et al. Programmed cell death 1 (PD-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol Biomarkers Prev 2014;23:2965–2970.
    1. Rosenberg SA, et al. 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;313:1485–1492.
    1. Escudier B, et al. Axitinib versus sorafenib in advanced renal cell carcinoma: Subanalyses by prior therapy from a randomised phase III trial. Br J Cancer 2014;110:2821–2828.
    1. Witte RS, et al. Eastern Cooperative Oncology Group phase II trial of ifosfamide in the treatment of previously treated advanced urothelial carcinoma. J Clin Oncol 1997;15:589–593.
    1. Lorusso V, et al. A phase II study of gemcitabine in patients with transitional cell carcinoma of the urinary tract previously treated with platinum. Italian Co-operative Group on Bladder Cancer. Eur J Cancer 1998;34:1208–1212.
    1. Bambury RM, et al. The safety and efficacy of single-agent pemetrexed in platinum-resistant advanced urothelial carcinoma: A large single-institution experience. Oncologist 2015;20:508–515.
    1. Gallagher DJ, et al. Phase II study of sunitinib in patients with metastatic urothelial cancer. J Clin Oncol 2010;28:1373–1379.
    1. Wulfing C, et al. A single-arm, multicenter, open-label phase 2 study of lapatinib as the second-line treatment of patients with locally advanced or metastatic transitional cell carcinoma. Cancer 2009;115:2881–2890.
    1. Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3–potential mechanisms of action. Nat Rev Immunol 2015;15:45–56.
    1. Rizvi N, et al. Optimizing PD-L1 as a biomarker of response with pembrolizumab (pembro; MK-3475) as first-line therapy for PD-L1–positive metastatic non-small cell lung cancer (NSCLC): Updated data from KEYNOTE-001. J Clin Oncol 2015;33(suppl); abstr 8026.
    1. Shankaran V, et al. Correlation of gene expression signatures and clinical outcomes in patients with advanced gastric cancer treated with pembrolizumab (MK-3475). J Clin Oncol 2015;33(suppl); abstr 3026.

Source: PubMed

3
구독하다