Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: a randomized clinical trial

F Stephen Hodi, Sandra Lee, David F McDermott, Uma N Rao, Lisa H Butterfield, Ahmad A Tarhini, Philip Leming, Igor Puzanov, Donghoon Shin, John M Kirkwood, F Stephen Hodi, Sandra Lee, David F McDermott, Uma N Rao, Lisa H Butterfield, Ahmad A Tarhini, Philip Leming, Igor Puzanov, Donghoon Shin, John M Kirkwood

Abstract

Importance: Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) blockade with ipilimumab prolongs survival in patients with metastatic melanoma. CTLA-4 blockade and granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor vaccine combinations demonstrate therapeutic synergy in preclinical models. A key unanswered question is whether systemic GM-CSF (sargramostim) enhances CTLA-4 blockade.

Objective: To compare the effect of ipilimumab plus sargramostim vs ipilimumab alone on overall survival (OS) in patients with metastatic melanoma.

Design, setting, and participants: The Eastern Cooperative Oncology Group (ECOG) conducted a US-based phase 2 randomized clinical trial from December 28, 2010, until July 28, 2011, of patients (N = 245) with unresectable stage III or IV melanoma, at least 1 prior therapy, no central nervous system metastases, and ECOG performance status of 0 or 1.

Interventions: Patients were randomized to receive ipilimumab, 10 mg/kg, intravenously on day 1 plus sargramostim, 250 μg subcutaneously, on days 1 to 14 of a 21-day cycle (n = 123) vs ipilimumab alone (n = 122). Ipilimumab treatment included induction for 4 cycles followed by maintenance every fourth cycle.

Main outcomes and measures: Primary end point: comparison of length of OS. Secondary end point: progression-free survival (PFS), response rate, safety, and tolerability.

Results: Median follow-up was 13.3 months (range, 0.03-19.9). Median OS as of December 2012 for ipilimumab plus sargramostim was 17.5 months (95% CI, 14.9-not reached) vs 12.7 months (95% CI, 10.0-not reached) for ipilimumab. The 1-year survival rate for ipilimumab plus sargramostim was 68.9% (95% CI, 60.6%-85.5%) compared to 52.9% (95% CI, 43.6%-62.2%) for ipilimumab alone (stratified log-rank 1-sided P = .01; mortality hazard ratio 0.64 [1-sided 90% repeated CI, not applicable-0.90]). A planned interim analysis was conducted at 69.8% of expected events (104 observed with 149 expected deaths). Planned interim analysis using the O'Brien-Fleming boundary was crossed for improvement in OS. There was no difference in PFS. Median PFS for ipilimumab plus sargramostim was 3.1 months (95% CI, 2.9-4.6) vs 3.1 months (95% CI, 2.9-4.0) for ipilimumab alone. Grade 3 to 5 adverse events occurred in 44.9% (95% CI; 35.8%-54.4%) of patients in the ipilimumab plus sargramostim group vs 58.3% (95% CI, 49.0%-67.2%) of patients in the ipilimumab-alone group (2-sided P = .04).

Conclusion and relevance: Among patients with unresectable stage III or IV melanoma, treatment with ipilimumab plus sargramostim vs ipilimumab alone resulted in longer OS and lower toxicity, but no difference in PFS. These findings require confirmation in larger studies with longer follow-up.

Trial registration: clinicaltrials.gov Identifier: NCT01134614.

Figures

Figure 1
Figure 1
Enrollment and Outcomes Diagram. There were 7 and 4 ineligible patients in Ipilimumab+Sargramostin and Ipilimumab treatment groups, respectively. These cases were confirmed to be ineligible based on the central review conducted at ECOG after randomization. All of them were included in the efficacy analysis.
Figure 2
Figure 2
Kaplan-Meier Estimates for overall survival and progression-free survival. Panel A shows the primary efficacy treatment analysis of sargramostim plus ipilimumab as compared to ipilimumab alone (stratified hazard ratio = 0.64; One-sided 90% Repeated Confidence Interval (RCI), with an upper bound 0.90; stratified log rank test one-sided P=.01). Panel B shows the Kaplan-Meier estimates for progression-free survival by treatment group in the intent-to-treat patient population. There was no significant difference in progression-free survival between treatment groups (stratified hazard ratio =.87, 95% confidence interval (CI), 0.64 , 1.18; stratified log rank test two-sided P=.37).
Figure 2
Figure 2
Kaplan-Meier Estimates for overall survival and progression-free survival. Panel A shows the primary efficacy treatment analysis of sargramostim plus ipilimumab as compared to ipilimumab alone (stratified hazard ratio = 0.64; One-sided 90% Repeated Confidence Interval (RCI), with an upper bound 0.90; stratified log rank test one-sided P=.01). Panel B shows the Kaplan-Meier estimates for progression-free survival by treatment group in the intent-to-treat patient population. There was no significant difference in progression-free survival between treatment groups (stratified hazard ratio =.87, 95% confidence interval (CI), 0.64 , 1.18; stratified log rank test two-sided P=.37).
Figure 3
Figure 3
Subgroup analyses for overall survival and progression-free survival in intent-to treat patient population. Panel A shows subgroup analyses of overall survival among subgroups of patients as defined by baseline characteristics (age, sex, ECOG PS, LDH) and stratification factors (prior therapy and metastasis (M) stage classified according to the tumor-none-metastasis (TNM) categorization for melanoma of the American Joint Committee on Cancer). The hazard ratios were lower than one indicating a lower risk of death in each subgroup in favor of the sargramostim plus ipilimumab group except for gender. There was a differential treatment effect on OS by gender. While male patients treated with Ipilimumab+Sargramostim had better overall survival, female patients treated with Ipilimumab alone had better overall survival. This trend needs to be interpreted with caution as the sample size and number of deaths in subgroups by gender were relatively small. Boxes represent hazard ratios, size of box inversely proportional to SE of HR. Bars represent two-sided 95% confidence intervals. The stratified HR for overall survival was .64 with one-sided 90% RCI (not applicable, .90). ECOG PS: Eastern Cooperative Oncology Group performance status, LDH: Lactate dehydrogenase. AJCC: American Joint Committee on Cancer. HRs and the 95%CI were calculated using univariate Cox models for each category. Overall HR was estimated from the Cox model stratified by AJCC stage and prior therapy. Panel B: Subgroup analyses of progression-free survival. There were no significant differences for progression-free survival by treatment in any of the sub-groups.
Figure 3
Figure 3
Subgroup analyses for overall survival and progression-free survival in intent-to treat patient population. Panel A shows subgroup analyses of overall survival among subgroups of patients as defined by baseline characteristics (age, sex, ECOG PS, LDH) and stratification factors (prior therapy and metastasis (M) stage classified according to the tumor-none-metastasis (TNM) categorization for melanoma of the American Joint Committee on Cancer). The hazard ratios were lower than one indicating a lower risk of death in each subgroup in favor of the sargramostim plus ipilimumab group except for gender. There was a differential treatment effect on OS by gender. While male patients treated with Ipilimumab+Sargramostim had better overall survival, female patients treated with Ipilimumab alone had better overall survival. This trend needs to be interpreted with caution as the sample size and number of deaths in subgroups by gender were relatively small. Boxes represent hazard ratios, size of box inversely proportional to SE of HR. Bars represent two-sided 95% confidence intervals. The stratified HR for overall survival was .64 with one-sided 90% RCI (not applicable, .90). ECOG PS: Eastern Cooperative Oncology Group performance status, LDH: Lactate dehydrogenase. AJCC: American Joint Committee on Cancer. HRs and the 95%CI were calculated using univariate Cox models for each category. Overall HR was estimated from the Cox model stratified by AJCC stage and prior therapy. Panel B: Subgroup analyses of progression-free survival. There were no significant differences for progression-free survival by treatment in any of the sub-groups.

References

    1. Inaba K, Inaba M, Romani N, et al. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med. 1992;176:1693–702.
    1. Fischer HG, Frosch S, Reske K, Reske-Kunz AB. Granulocyte-macrophage colony-stimulating factor activates macrophages derived from bone marrow cultures to synthesis of MHC class II molecules and to augmented antigen presentation function. J Immunol. 1988;141:3882–8.
    1. Weisbart RH, Golde DW, Clark SC, Wong GG, Gasson JC. Human granulocyte-macrophage colony-stimulating factor is a neutrophil activator. Nature. 1985;314:361–3.
    1. Small EJ, Reese DM, Um B, Whisenant S, Dixon SC, Figg WD. Therapy of advanced prostate cancer with granulocyte macrophage colony-stimulating factor. Clin Cancer Res. 1999;5:1738–44.
    1. Everly JJ, Lonial S. Immunomodulatory effects of human recombinant granulocyte-macrophage colony-stimulating factor (rhuGM-CSF): evidence of antitumour activity. Expert Opin Biol Ther. 2005;5:293–311.
    1. Slingluff CL, Jr., Petroni GR, Olson WC, et al. Effect of granulocyte/macrophage colony-stimulating factor on circulating CD8+ and CD4+ T-cell responses to a multipeptide melanoma vaccine: outcome of a multicenter randomized trial. Clin Cancer Res. 2009;15:7036–44.
    1. Hodi FS, O'Day SJ, McDermott DF, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med. 2010;363:711–23.
    1. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 364:2517–26.
    1. van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med. 1999;190:355–66.
    1. Hodi FS, Mihm MC, Soiffer RJ, et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A. 2003;100:4712–7.
    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;105:3005–10.
    1. van den Eertwegh AJ, Versluis J, van den Berg HP, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. The lancet oncology. 2012;13:509–17.
    1. Fong L, Kwek SS, O'Brien S, et al. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 2009;69:609–15.
    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;15:7412–20.
    1. O'Brien PC, Fleming TR. A multiple testing procedure for clinical trials. Biometrics. 1979;35:549–56.
    1. Jennison C, Turnbull BW. Group Sequential Analysis Incorporating Covariate Information. Journal of the American Statistical Association. 1997;92:1330–41.
    1. Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res. 2011;71:5445–54.
    1. Tang D, Shen Y, Sun J, et al. Increased frequency of ICOS+ CD4 T-cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Re. 2013;1:1–6.
    1. Kaplan EL, Meier P. Nonparametric estimation from incomplete observation. Journal of Americak Statistical Association. 1958;53:457–81.
    1. Cox DR. Regression models and life tables (with discussion). Journal of the Royal Statistical Society Series B. 1972;34:187–220.
    1. Agresti A. Cataegorical Data Analysis. Wiley; New York: 1990.
    1. Korn EL, Liu PY, Lee SJ, et al. Meta-analysis of phase II cooperative group trials in metastatic stage IV melanoma to determine progression-free and overall survival benchmarks for future phase II trials. J Clin Oncol. 2008;26:527–34.
    1. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22.
    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. Jinushi M, Nakazaki Y, Dougan M, Carrasco DR, Mihm M, Dranoff G. MFG-E8-mediated uptake of apoptotic cells by APCs links the pro- and antiinflammatory activities of GM-CSF. J Clin Invest. 2007;117:1902–13.
    1. Filipazzi P, Valenti R, Huber V, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol. 2007;25:2546–53.
    1. Parmiani G, Castelli C, Pilla L, Santinami M, Colombo MP, Rivoltini L. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann Oncol. 2007;18:226–32.
    1. Sotomayor EM, Fu YX, Lopez-Cepero M, et al. Role of tumor-derived cytokines on the immune system of mice bearing a mammary adenocarcinoma. II. Down-regulation of macrophage-mediated cytotoxicity by tumor-derived granulocyte-macrophage colony-stimulating factor. J Immunol. 1991;147:2816–23.
    1. Selby MJ, Engelhardt JJ, Quigley M, et al. Anti-CTLA-4 Antibodies of IgG2a Isotype Enhance Antitumor Activity through Reduction of Intratumoral Regulatory T Cells. Cancer Immunology Research. 2013;1:29–39.
    1. Hercus TR, Thomas D, Guthridge MA, et al. The granulocyte-macrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease. Blood. 2009;114:1289–98.
    1. Han X, Uchida K, Jurickova I, et al. Granulocyte-macrophage colony-stimulating factor autoantibodies in murine ileitis and progressive ileal Crohn's disease. Gastroenterology. 2009;136:1261–71. e1–3.
    1. Goldstein JI, Kominsky DJ, Jacobson N, et al. Defective leukocyte GM-CSF receptor (CD116) expression and function in inflammatory bowel disease. Gastroenterology. 2011;141:208–16.
    1. Dranoff G. Granulocyte-macrophage colony stimulating factor and inflammatory bowel disease: establishing a connection. Gastroenterology. 2011;141:28–31.
    1. Egea L, Hirata Y, Kagnoff MF. GM-CSF: a role in immune and inflammatory reactions in the intestine. Expert review of gastroenterology & hepatology. 2010;4:723–31.
    1. Xu Y, Hunt NH, Bao S. The role of granulocyte macrophage-colony-stimulating factor in acute intestinal inflammation. Cell research. 2008;18:1220–9.
    1. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007;204:1757–64.
    1. Fukuzawa H, Sawada M, Kayahara T, et al. Identification of GM-CSF in Paneth cells using single-cell RT-PCR. Biochem Biophys Res Commun. 2003;312:897–902.
    1. Dranoff G, Crawford AD, Sadelain M, et al. Involvement of granulocyte-macrophage colony-stimulating factor in pulmonary homeostasis. Science. 1994;264:713–6.
    1. Soroosh P, Doherty TA, Duan W, et al. Lung-resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J Exp Med. 2013 Epub head of print.
    1. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366:2443–54.

Source: PubMed

3
Prenumerera