Human Epidermal Growth Factor Receptor 2 (HER2) -Specific Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma.

PURPOSE The outcome for patients with metastatic or recurrent sarcoma remains poor. Adoptive therapy with tumor-directed T cells is an attractive therapeutic option but has never been evaluated in sarcoma. PATIENTS AND METHODS We conducted a phase I/II clinical study in which patients with recurrent/refractory human epidermal growth factor receptor 2 (HER2) -positive sarcoma received escalating doses (1 × 10(4)/m(2) to 1 × 10(8)/m(2)) of T cells expressing an HER2-specific chimeric antigen receptor with a CD28.ζ signaling domain (HER2-CAR T cells). RESULTS We enrolled 19 patients with HER2-positive tumors (16 osteosarcomas, one Ewing sarcoma, one primitive neuroectodermal tumor, and one desmoplastic small round cell tumor). HER2-CAR T-cell infusions were well tolerated with no dose-limiting toxicity. At dose level 3 (1 × 10(5)/m(2)) and above, we detected HER2-CAR T cells 3 hours after infusion by quantitative polymerase chain reaction in 14 of 16 patients. HER2-CAR T cells persisted for at least 6 weeks in seven of the nine evaluable patients who received greater than 1 × 10(6)/m(2) HER2-CAR T cells (P = .005). HER2-CAR T cells were detected at tumor sites of two of two patients examined. Of 17 evaluable patients, four had stable disease for 12 weeks to 14 months. Three of these patients had their tumor removed, with one showing ≥ 90% necrosis. The median overall survival of all 19 infused patients was 10.3 months (range, 5.1 to 29.1 months). CONCLUSION This first evaluation of the safety and efficacy of HER2-CAR T cells in patients with cancer shows the cells can persist for 6 weeks without evident toxicities, setting the stage for studies that combine HER2-CAR T cells with other immunomodulatory approaches to enhance their expansion and persistence.

[1]  E. Kleinerman,et al.  Genetically modified T cells targeting interleukin-11 receptor α-chain kill human osteosarcoma cells and induce the regression of established osteosarcoma lung metastases. , 2012, Cancer research.

[2]  M. Fishman,et al.  Cellular immunotherapy for soft tissue sarcomas. , 2012, Immunotherapy.

[3]  C. Rooney,et al.  Enhanced Tumor Trafficking of GD2 Chimeric Antigen Receptor T Cells by Expression of the Chemokine Receptor CCR2b , 2010, Journal of immunotherapy.

[4]  S. Gottschalk,et al.  Immunotherapy targeting HER2 with genetically modified T cells eliminates tumor-initiating cells in osteosarcoma , 2011, Cancer Gene Therapy.

[5]  M. van Glabbeke,et al.  New guidelines to evaluate the response to treatment in solid tumors , 2000, Journal of the National Cancer Institute.

[6]  David L. Porter,et al.  T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia , 2011, Science Translational Medicine.

[7]  Michel Sadelain,et al.  PD-1– and CTLA-4–Based Inhibitory Chimeric Antigen Receptors (iCARs) Divert Off-Target Immunotherapy Responses , 2013, Science Translational Medicine.

[8]  Sylvia Janetzki,et al.  Improved Endpoints for Cancer Immunotherapy Trials , 2010, Journal of the National Cancer Institute.

[9]  D. Busch,et al.  Adoptive transfer of autologous, HER2-specific, cytotoxic T lymphocytes for the treatment of HER2-overexpressing breast cancer , 2007, Cancer Immunology, Immunotherapy.

[10]  Qing He,et al.  CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia , 2013, Science Translational Medicine.

[11]  J. Waisman,et al.  Phase I study of infusion of HER2/neu (HER2) specific T cells in patients with advanced-stage HER2 overexpressing cancers who have received a HER2 vaccine. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[12]  Adrian P Gee,et al.  Inducible apoptosis as a safety switch for adoptive cell therapy. , 2011, The New England journal of medicine.

[13]  S. Gottschalk,et al.  Design and development of therapies using chimeric antigen receptor‐expressing T cells , 2014, Immunological reviews.

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

[15]  B. Gerstmayer,et al.  Costimulation of T-cell proliferation by a chimeric B7-antibody fusion protein , 1997, Cancer Immunology, Immunotherapy.

[16]  Hao Liu,et al.  CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. , 2011, The Journal of clinical investigation.

[17]  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.

[18]  S. Ferrari,et al.  Treatment and outcome of recurrent osteosarcoma: Experience at Rizzoli in 235 patients initially treated with neoadjuvant chemotherapy , 2005, Acta oncologica.

[19]  M Van Glabbeke,et al.  New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. , 2000, Journal of the National Cancer Institute.

[20]  S. Rosenberg,et al.  Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[21]  Bernd Hauck,et al.  Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. , 2013, The New England journal of medicine.

[22]  P. Meyers High-dose therapy with autologous stem cell rescue for pediatric sarcomas , 2004, Current opinion in oncology.

[23]  R. Gorlick,et al.  Ganglioside GD2 as a therapeutic target for antibody‐mediated therapy in patients with osteosarcoma , 2014, Cancer.

[24]  W. Wilson,et al.  B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. , 2012, Blood.

[25]  S. Lipshultz,et al.  Phase II trial of trastuzumab in combination with cytotoxic chemotherapy for treatment of metastatic osteosarcoma with human epidermal growth factor receptor 2 overexpression: a report from the children's oncology group. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[26]  J. Squire,et al.  HER2 Amplification and Overexpression Is Not Present in Pediatric Osteosarcoma: A Tissue Microarray Study , 2005, Pediatric and developmental pathology : the official journal of the Society for Pediatric Pathology and the Paediatric Pathology Society.

[27]  M. Schilham,et al.  Chemotherapy-resistant osteosarcoma is highly susceptible to IL-15-activated allogeneic and autologous NK cells , 2011 .

[28]  A. M. Stanley,et al.  Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab , 2003, Nature.

[29]  Khin Thway,et al.  Systemic treatment of soft-tissue sarcoma—gold standard and novel therapies , 2014, Nature Reviews Clinical Oncology.

[30]  S. Gultekin,et al.  Monoclonal antibody 8H9 targets a novel cell surface antigen expressed by a wide spectrum of human solid tumors. , 2001, Cancer research.

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

[32]  Martin Pule,et al.  Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. , 2011, Blood.

[33]  M. Semik,et al.  Osteosarcoma relapse after combined modality therapy: an analysis of unselected patients in the Cooperative Osteosarcoma Study Group (COSS). , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[34]  M. Slovak,et al.  Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. , 2007, Molecular therapy : the journal of the American Society of Gene Therapy.

[35]  R. Grossman,et al.  HER2-Specific T Cells Target Primary Glioblastoma Stem Cells and Induce Regression of Autologous Experimental Tumors , 2010, Clinical Cancer Research.

[36]  K. Blum,et al.  Lymphocyte numbers and subsets in the human blood. Do they mirror the situation in all organs? , 2007, Immunology letters.

[37]  S. Bielack,et al.  Bone Tumors in Adolescents and Young Adults , 2008, Current treatment options in oncology.

[38]  M. Kalos,et al.  Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. , 2010, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[39]  A. Huvos,et al.  Expression of HER2/erbB-2 correlates with survival in osteosarcoma. , 1999, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[40]  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.

[41]  C. Beauchamp Second and Subsequent Recurrences of Osteosarcoma: Presentation, Treatment, and Outcomes of 249 Consecutive Cooperative Osteosarcoma Study Group Patients , 2009 .

[42]  D. Campana,et al.  Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[43]  A. Bagg,et al.  Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. , 2011, The New England journal of medicine.