Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma.

We generated MHC-independent chimeric antigen receptors (CARs) directed to the GD2 antigen expressed by neuroblastoma tumor cells and treated patients with this disease. Two distinguishable forms of this CAR were expressed in EBV-specific cytotoxic T lymphocytes (EBV-CTLs) and activated T cells (ATCs). We have previously shown that EBV-CTLs expressing GD2-CARs (CAR-CTLs) circulated at higher levels than GD2-CAR ATCs (CAR-ATCs) early after infusion, but by 6 weeks, both subsets became low or undetectable. We now report the long-term clinical and immunologic consequences of infusions in 19 patients with high-risk neuroblastoma: 8 in remission at infusion and 11 with active disease. Three of 11 patients with active disease achieved complete remission, and persistence of either CAR-ATCs or CAR-CTLs beyond 6 weeks was associated with superior clinical outcome. We observed persistence for up to 192 weeks for CAR-ATCs and 96 weeks for CAR-CTLs, and duration of persistence was highly concordant with the percentage of CD4(+) cells and central memory cells (CD45RO(+)CD62L(+)) in the infused product. In conclusion, GD2-CAR T cells can induce complete tumor responses in patients with active neuroblastoma; these CAR T cells may have extended, low-level persistence in patients, and such persistence was associated with longer survival. This study is registered at www.clinialtrials.gov as #NCT00085930.

[1]  S. Riddell,et al.  Engraftment of human central memory-derived effector CD8+ T cells in immunodeficient mice. , 2011, Blood.

[2]  J. Trapani,et al.  Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors. , 2002, Blood.

[3]  K. Matthay,et al.  A Phase I/IB trial of murine monoclonal anti‐GD2 antibody 14.G2a plus interleukin‐2 in children with refractory neuroblastoma , 1997, Cancer.

[4]  P. Reuland,et al.  A phase I study of neuroblastoma with the anti-ganglioside GD2 antibody 14.G2a , 2005, Cancer Immunology, Immunotherapy.

[5]  Helen X. Chen,et al.  Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. , 2010, The New England journal of medicine.

[6]  S. Ferrone,et al.  Mechanisms of immune evasion of human neuroblastoma. , 2005, Cancer letters.

[7]  D. Srivastava,et al.  Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. , 1998, Blood.

[8]  F. Berthold,et al.  Expression of MHC class I, MHC class II, and cancer germline antigens in neuroblastoma , 2005, Cancer Immunology, Immunotherapy.

[9]  M. Brenner,et al.  Cellular immunotherapy for neuroblastoma: a review of current vaccine and adoptive T cell therapeutics. , 2009, Current pharmaceutical design.

[10]  C. Rooney,et al.  Target Antigen Expression on a Professional Antigen-Presenting Cell Induces Superior Proliferative Antitumor T-Cell Responses via Chimeric T-Cell Receptors , 2006, Journal of immunotherapy.

[11]  R. Gilbertson,et al.  Regression of experimental medulloblastoma following transfer of HER2-specific T cells. , 2007, Cancer research.

[12]  J. Murray,et al.  Phase I trial of murine monoclonal antibody 14G2a administered by prolonged intravenous infusion in patients with neuroectodermal tumors. , 1994, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[13]  H. Heslop,et al.  Immunotherapy for osteosarcoma: genetic modification of T cells overcomes low levels of tumor antigen expression. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[14]  R. Gray A Class of $K$-Sample Tests for Comparing the Cumulative Incidence of a Competing Risk , 1988 .

[15]  N. Berger,et al.  Ganglioside GD2 specific monoclonal antibody 3F8: a phase I study in patients with neuroblastoma and malignant melanoma. , 1987, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[16]  M. Schrappe,et al.  A phase I study of human/mouse chimeric antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. , 1995, European journal of cancer.

[17]  S. Ferrone,et al.  Immunogenicity of Human Neuroblastoma , 2004, Annals of the New York Academy of Sciences.

[18]  D. Neuberg,et al.  Long-Lived Antitumor CD8+ Lymphocytes for Adoptive Therapy Generated Using an Artificial Antigen-Presenting Cell , 2007, Clinical Cancer Research.

[19]  Mike Gough,et al.  Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. , 2008, The Journal of clinical investigation.

[20]  F. Berthold,et al.  Consolidation treatment with chimeric anti-GD2-antibody ch14.18 in children older than 1 year with metastatic neuroblastoma. , 2004, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  K. Matthay,et al.  A Phase I Clinical Trial of the hu14.18-IL2 (EMD 273063) as a Treatment for Children with Refractory or Recurrent Neuroblastoma and Melanoma: a Study of the Children's Oncology Group , 2006, Clinical Cancer Research.

[22]  Sylvain Julien,et al.  Retargeting of Human T Cells to Tumor-Associated MUC1: The Evolution of a Chimeric Antigen Receptor1 , 2008, The Journal of Immunology.

[23]  H. Heslop,et al.  Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease. , 2007, Blood.

[24]  P. Sperryn,et al.  Blood. , 1989, British journal of sports medicine.

[25]  J Pritchard,et al.  Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. , 1993, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[26]  M. Raffeld,et al.  Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes , 2002, Science.

[27]  B. Kushner,et al.  Phase II trial of the anti-G(D2) monoclonal antibody 3F8 and granulocyte-macrophage colony-stimulating factor for neuroblastoma. , 2001, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  C. Rooney,et al.  Improving T cell therapy for cancer. , 2007, Expert opinion on biological therapy.

[29]  A. Temme,et al.  Chimeric Antigen Receptor-Engineered T Cells for Immunotherapy of Cancer , 2010, Journal of biomedicine & biotechnology.

[30]  Egbert Oosterwijk,et al.  Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. , 2011, Blood.

[31]  K. Matthay,et al.  Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: a Children's Cancer Group Study. , 2000, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[32]  L. Sorkin,et al.  Anti-GD2 with an FC point mutation reduces complement fixation and decreases antibody-induced allodynia , 2010, PAIN®.

[33]  Michel Sadelain,et al.  The promise and potential pitfalls of chimeric antigen receptors. , 2009, Current Opinion in Immunology.

[34]  F. Berthold,et al.  Long term outcome of high-risk neuroblastoma patients after immunotherapy with antibody ch14.18 or oral metronomic chemotherapy , 2011, BMC Cancer.

[35]  H. Heslop,et al.  A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. , 2005, Molecular therapy : the journal of the American Society of Gene Therapy.

[36]  C. Rooney,et al.  Epstein-Barr virus-specific human T lymphocytes expressing antitumor chimeric T-cell receptors: potential for improved immunotherapy. , 2002, Blood.

[37]  Michel Sadelain,et al.  Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ /CD28 receptor , 2002, Nature Biotechnology.

[38]  Hao Liu,et al.  Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma , 2008, Nature Medicine.

[39]  S. Rosenberg,et al.  Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. , 2010, Blood.