Improved endpoints for cancer immunotherapy trials

Axel Hoos, Alexander M M Eggermont, Sylvia Janetzki, F Stephen Hodi, Ramy Ibrahim, Aparna Anderson, Rachel Humphrey, Brent Blumenstein, Lloyd Old, Jedd Wolchok, Axel Hoos, Alexander M M Eggermont, Sylvia Janetzki, F Stephen Hodi, Ramy Ibrahim, Aparna Anderson, Rachel Humphrey, Brent Blumenstein, Lloyd Old, Jedd Wolchok

Abstract

Unlike chemotherapy, which acts directly on the tumor, cancer immunotherapies exert their effects on the immune system and demonstrate new kinetics that involve building a cellular immune response, followed by changes in tumor burden or patient survival. Thus, adequate design and evaluation of some immunotherapy clinical trials require a new development paradigm that includes reconsideration of established endpoints. Between 2004 and 2009, several initiatives facilitated by the Cancer Immunotherapy Consortium of the Cancer Research Institute and partner organizations systematically evaluated an immunotherapy-focused clinical development paradigm and created the principles for redefining trial endpoints. On this basis, a body of clinical and laboratory data was generated that supports three novel endpoint recommendations. First, cellular immune response assays generate highly variable results. Assay harmonization in multicenter trials may minimize variability and help to establish cellular immune response as a reproducible biomarker, thus allowing investigation of its relationship with clinical outcomes. Second, immunotherapy may induce novel patterns of antitumor response not captured by Response Evaluation Criteria in Solid Tumors or World Health Organization criteria. New immune-related response criteria were defined to more comprehensively capture all response patterns. Third, delayed separation of Kaplan-Meier curves in randomized immunotherapy trials can affect results. Altered statistical models describing hazard ratios as a function of time and recognizing differences before and after separation of curves may allow improved planning of phase III trials. These recommendations may improve our tools for cancer immunotherapy trials and may offer a more realistic and useful model for clinical investigation.

Figures

Figure 1
Figure 1
High variability of results for the enzyme-linked immunosorbent spot (ELISPOT) immune response assay. Identical peripheral blood mononuclear cell samples from the same patient were sent to 36 different laboratories experienced with ELISPOT methodology. The image shows the spot count results in microtiter plates in which each well represents the result of one laboratory. Some wells show high numbers of spots, whereas others are low or negative. Each spot in this assay represents a single T-cell capable of reacting against a defined target antigen. These results reflect the outcome of the first ELISPOT proficiency panel, which identified sources of variability among laboratories.
Figure 2
Figure 2
Effect of assay harmonization on data variability of the enzyme-linked immunosorbent spot (ELISPOT) assay. In the Cancer Immunotherapy Consortium of the Cancer Research Institute ELISPOT proficiency panel, participating laboratories reported the response status from eight different donor–antigen combinations. Grey bars represent the first panel round and stippled bars the second panel round. In the first panel round, 47% of panelists missed detection of at least one response correctly, and 11% of panelists failed to detect at least three responses correctly (characterized as an “outlier” because of high variability). Based on the first panel results, harmonization criteria were given to panelists, and the testing was repeated in the second panel (stippled bars). ELISPOT performance improved, with only 14% of panelists missing at least one responder and zero outliers.
Figure 3
Figure 3
Clinical response patterns observed with anti-cytotoxic T lymphocyte–associated protein 4 immunotherapy (ipilimumab). Immunotherapy patterns of response depicted as a continuous variable of relative change of tumor burden (%) over time. Tumor burden is described through the sum of the perpendicular diameters (SPD) of all measurable lesions (baseline and new) at each time point. A and B) Conventional response patterns: (A) immediate response; (B) durable stable disease with possible slow decline in tumor burden. C and E) Novel immunotherapy response patterns: (C) increase in total tumor burden followed by response. (D) Clinical images corresponding to pattern (C): tumor burden on the skin at baseline (day 0) is increased at first follow-up (day 84) and subsequently declines (day 112) to a complete response (day 503) (courtesy of Dr K. Harmankaya). E) The second novel pattern shows a response in the presence of new lesions; existing lesions present at baseline (blue) and new lesions (red) are added to define the total tumor burden (green). Despite new lesions, the total tumor burden is still declining to a partial response. Yellow triangles indicate dosing with immunotherapy; horizontal lines indicate standard thresholds for response or progression. Modified after Wolchok et al. (43).
Figure 4
Figure 4
Delayed separation of survival curves of sipuleucel-T immunotherapy vs placebo in advanced prostate cancer, where the separation of Kaplan–Meier curves occurred after approximately 8 months after random assignment. HR = hazard ratio; CI = confidence interval. Courtesy of Small et al. (12). Reprinted with permission. Copyright 2008 American Society of Clinical Oncology. All rights reserved.
Figure 5
Figure 5
Mathematical illustration of a delayed separation of curves. Example of a two-arm study with an ultimate hazard ratio of 0.7. The control arm has an exponential survival distribution with median survival of 18 months (red dashed curve). The form of the delayed separation is specified by a hazard ratio function (HR(t), solid gray line) that has the value 1.0 for 3 months and then decreases linearly between 3 and 6 months to become 0.7 at 6 months and then remains constant. The experimental arm survival distribution (solid blue line) is the consequence of mathematically blending the control arm survival distribution function and the hazard ratio function.

References

    1. Finke LH, Wentworth K, Blumenstein B, et al. Lessons from randomized phase III studies with active cancer immunotherapies—outcomes from the 2006 meeting of the Cancer Vaccine Consortium (CVC) Vaccine. 2007;25(suppl 2):B97–B109.
    1. Copier J, Dalgleish AG, Britten CM, et al. Improving the efficacy of cancer immunotherapy. Eur J Cancer. 2009;45(8):1424–1431.
    1. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10(9):909–915.
    1. Finn OJ. Cancer immunology. N Engl J Med. 2008;358(25):2704–2715.
    1. Melero I, Hervas-Stubbs S, Glennie M, et al. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer. 2007;7(2):95–106.
    1. Peggs KS, Quezada SA, Korman AJ, et al. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Curr Opin Immunol. 2006;18(2):206–213.
    1. Ribas A, Butterfield LH, Glaspy JA, et al. Current developments in cancer vaccines and cellular immunotherapy. J Clin Oncol. 2003;21(12):2415–2432.
    1. Hoos A, Parmiani G, Hege K, et al. A clinical development paradigm for cancer vaccines and related biologics. J Immunother. 2007;30(1):1–15.
    1. Hoos A. Proposal of a clinical development paradigm for cancer immunotherapy: novel endpoints. In: Endpoints for Immunotherapy Studies: Design and Regulatory Implications, American Society of Clinical Oncology (ASCO) Annual Meeting; June 2, 2008; Chicago, IL.
    1. Michaelis LC, Ratain MJ. Measuring response in a post-RECIST world: from black and white to shades of grey. Nat Reviews Cancer. 2006;6(5):409–414.
    1. Bendandi M. Idiotype vaccines for lymphoma: proof-of-principles and clinical trial failures. Nat Reviews Cancer. 2009;9(9):675–681.
    1. Small EJ, Schellhammer PF, Higano CS. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24(19):3089–3094.
    1. Small EJ, Fong L. Developing immunotherapy as legitimate therapy for patients with prostate cancer [published online ahead of print January 25, 2010] J Clin Oncol. 2010;28(7):1085–1087.
    1. Kantoff PW, Schuetz TJ, Blumenstein BA, et al. Overall survival analysis of a phase II randomized controlled trial of a poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer [published online ahead of print January 25, 2010] J Clin Oncol. 2010;28(7):1099–1105.
    1. Hamid O, Urba WJ, Yellin M, et al. Kinetics of response to ipilimumab (MDX-010) in patients with stage III/IV melanoma [abstract 8525] J Clin Oncol. 2007;25(suppl):18s.
    1. Wolchok JD, Ibrahim R, DePril V, et al. Antitumor response and new lesions in advanced melanoma patients on ipilimumab treatment [abstract 3020] J Clin Oncol. 2008;26(suppl):19s.
    1. Wolchok JD, Neyns B, Linette G, et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol. 2010;11(2):155–164.
    1. Inoges S, Rodriguez-Calvillo M, Zabalegui N, et al. Clinical benefit associated with idiotype vaccination in patients with follicular lymphoma. J Natl Cancer Inst. 2006;98(18):1292–1301.
    1. Longo DL. Idiotype vaccination in follicular lymphoma: knocking on the doorway to cure. J Natl Cancer Inst. 2006;98(18):1263–1265.
    1. Janetzki S, Panageas KS, Ben-Porat L, et al. for the Elispot Proficiency Panel of the CVC Immune Assay Working Group 2007. Results and Harmonization Guidelines from two large-scale international Elispot proficiency panels conducted by the Cancer Vaccine Consortium (CVC/SVI) Cancer Immunol Immunother. 2008;57(3):303–315.
    1. Britten CM, Gouttefangeas C, Schoenmaekers-Welters MJP, et al. The CIMT-Monitoring panel: a two-step approach to harmonize the enumeration of antigen-specific CD8+ T lymphocytes by structural and functional assays. Cancer Immunol Immunother. 2008;57(3):303–315.
    1. Britten CM, Janetzki S, Ben-Porat L, et al. for the HLA-peptide Multimer Proficiency Panel of the CVC-CRI Immune Assay Working Group. Harmonization guidelines for HLA-peptide multimer assays derived from results of a large scale international proficiency panel of the Cancer Vaccine Consortium (CVC) Cancer Immunol Immunother. 2009;58(10):1701–1713.
    1. Guidance for Industry: Clinical Considerations for Therapeutic Cancer Vaccines. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Biologics Evaluation and Research; 2009. .
    1. Maecker HT. The role of immune monitoring in evaluating cancer immunotherapy. In: Disis ML, editor. Cancer Drug Discovery and Development: Immunotherapy of Cancer. Totowa, NJ: Humana Press; 2005. pp. 59–72.
    1. Hobeika AC, Morse MA, Osada T, et al. Enumerating antigen-specific T-cell responses in peripheral blood: a comparison of peptide MHC Tetramer, ELISpot, and intracellular cytokine analysis. J Immunother. 2005;28(1):63–72.
    1. Walker EB, Disis ML. Monitoring immune responses in cancer patients receiving tumor vaccines. Int Rev Immunol. 2003;22(3–4):283–319.
    1. Keilholz U, Weber J, Finke LH, et al. Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the Society for Biological Therapy. J Immunother. 2002;25(2):97–138.
    1. Janetzki S, Britten CM, Kalos M, et al. MIATA—minimal information about T cell assays. Immunity. 2009;31(4):527–528.
    1. Tuomela M, Stanescu I, Krohn K. Validation overview of bio-analytical methods. Gene Ther. 2005;12(suppl 1):S131–S138.
    1. Britten CM, Janetzki S, van der Burg SH, et al. Toward the harmonization of immune monitoring in clinical trials: quo vadis? Cancer Immunol Immunother. 2008;57(3):285–288.
    1. International Conference on Harmonization (ICH) Guidance for Industry: Good Clinical Practice. 1997;62(90):25691–25709E7.
    1. NCCLS. Evaluation of Matrix Effects, Approved Guidelines. NCCLS document EP14-A [ISBN 1-56238- 434-1]. Wayne, PA: NCCLS
    1. Janetzki S, Price L, Britten CM, et al. Performance of serum-supplemented and serum-free media in IFNgamma Elispot Assays for human T cells. Cancer Immunol Immunother. 2010;59(4):609–618.
    1. WHO Handbook for Reporting Results of Cancer Treatment. Geneva, Switzerland: World Health Organization Offset Publication No. 48; 1979.
    1. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst. 2000;92(3):205–216.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247.
    1. Berd D, Sato T, Cohn H, et al. Treatment of metastatic melanoma with autologous, hapten-modified melanoma vaccine: regression of pulmonary metastases. Int J Cancer. 2001;94(4):531–539.
    1. Kruit WHJ, van Ojik HH, Brichard VG, et al. Phase 1/2 study of subcutaneous and intradermal immunization with a recombinant MAGE-3 protein in patients with detectable metastatic melanoma. Int J Cancer. 2005;117(4):596–604.
    1. van Baren N, Bonnet MC, Dréno B, et al. Tumoral and immunologic response after vaccination of melanoma patients with an ALVAC virus encoding MAGE antigens recognized by T cells. J Clin Oncol. 2005;23(35):9008–9021.
    1. Hodi FS, Butler M, Oble DA, et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A. 2008;105(8):3005–3010.
    1. Weber J, Thompson JA, Hamid O, et al. A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin Cancer Res. 2009;15(17):5591–5598.
    1. O'Day S, Ibrahim R, DePril V, et al. Efficacy and safety of ipilimumab induction and maintenance dosing in patients with advanced melanoma who progressed on one or more prior therapies [abstract 9021] Proc Am Soc Clin Onc. 2008;26(20 suppl)
    1. Wolchok JD, Hoos A, O'Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15(23):7412–7420.
    1. Hodi FS, Hoos A, Ibrahim R, et al. Novel efficacy criteria for antitumor activity to immunotherapy using the example of ipilimumab, and anti-CTLA-4 monoclonal antibody [abstract 3008] J Clin Oncol. 2008;26(suppl):19s.
    1. Tuma RS. New response criteria proposed for immunotherapies. J Natl Cancer Inst. 2008;100(18):1280–1281.
    1. Sosman JA, Unger JM, Liu PY, et al. Southwest Oncology Group. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol. 2002;20(8):2067–2075.
    1. Testori A, Richards J, Whitman E, et al. for the C-100-21 Study Group. Comparison of autologous tumor-derived heat shock protein gp96-peptide complexes (vitespen) and physician's choice in a randomized phase 3 trial in patients with stage IV melanoma. J Clin Oncol. 2008;26(6):955–962.
    1. Wood C, Srivastava P, Bukowski R, et al. for the C-100-12 RCC Study Group. An adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: a multicentre, open-label, randomised phase III trial. Lancet. 2008;372(9633):145–154.
    1. Hodi FS, O'Day S, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma [published online ahead of print June 14, 2010] N Engl J Med. 2010 doi:10.1056/NEJMoa1003466.
    1. Fine GD. Consequences of delayed treatment effects on analysis of time-to-event endpoints. Drug Inf J. 2007;41:535–539.
    1. Fleming TR, Harrington DP. Counting Processes and Survival Analysis. New York, NY: John Wiley & Sons; 1991.
    1. Lakatos E. Sample size based on the log-rank statistic in complex clinical trials. Biometrics. 1988;44(1):229–241.
    1. Sposto R, Stablein D, Carter-Campbell S. A partially grouped logrank test. Stat Med. 1997;16(6):695–704.
    1. Karrison TG, Maitland ML, Stadler WM, et al. Design of phase II cancer trials using a continuous endpoint of change in tumor size: application to a study of sorafenib and erlotinib in non-small-cell lung cancer. J Natl Cancer Inst. 2007;99(19):1455–1461.
    1. Hodi FS, Oble DA, Drappatz J, et al. CTLA-4 blockade with ipilimumab induces significant clinical benefit in a female with melanoma metastases to the CNS. Nat Clin Pract Oncol. 2008;5(9):557–561.

Source: PubMed

3
Se inscrever